(19)
(11) EP 2 568 979 B1

(12) EUROPEAN PATENT SPECIFICATION

(45) Mention of the grant of the patent:
05.04.2017 Bulletin 2017/14

(21) Application number: 11781133.1

(22) Date of filing: 10.05.2011
(51) International Patent Classification (IPC): 
A61K 31/337(2006.01)
A61K 47/30(2006.01)
A61K 9/16(2006.01)
A61K 47/50(2017.01)
A61K 9/24(2006.01)
A61P 35/00(2006.01)
(86) International application number:
PCT/US2011/035903
(87) International publication number:
WO 2011/143201 (17.11.2011 Gazette 2011/46)

(54)

RATIOMETRIC COMBINATORIAL DRUG DELIVERY

RATIOMETRISCHE KOMBINATORISCHE ARZNEIMITTELVERABREICHUNG

ADMINISTRATION DE MÉDICAMENTS COMBINATOIRE RATIOMÉTRIQUE


(84) Designated Contracting States:
AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

(30) Priority: 10.05.2010 US 333138 P

(43) Date of publication of application:
20.03.2013 Bulletin 2013/12

(73) Proprietor: The Regents of the University of California
Oakland, CA 94607-5200 (US)

(72) Inventors:
  • ZHANG, Liangfang
    San Diego CA 62130 (US)
  • ARYAL, Santosh
    San Diego CA 92122 (US)
  • HU, Che-ming
    La Jolla CA 92092 (US)

(74) Representative: Weickmann & Weickmann PartmbB 
Postfach 860 820
81635 München
81635 München (DE)


(56) References cited: : 
WO-A1-2008/124632
US-A1- 2003 147 945
WO-A2-2008/051565
US-A1- 2006 046 967
   
  • SONG ET AL: "PLGA nanoparticles simultaneously loaded with vincristine sulfate and verapamil hydrochloride: Systematic study of particle size and drug entrapment efficiency", INTERNATIONAL JOURNAL OF PHARMACEUTICS, ELSEVIER BV, NL, vol. 350, no. 1-2, 28 January 2008 (2008-01-28), pages 320-329, XP022435790, ISSN: 0378-5173, DOI: 10.1016/J.IJPHARM.2007.08.034
  • LAMMERS T ET AL: "Simultaneous delivery of doxorubicin and gemcitabine to tumors in vivo using prototypic polymeric drug carriers", BIOMATERIALS, ELSEVIER SCIENCE PUBLISHERS BV., BARKING, GB, vol. 30, no. 20, 1 July 2009 (2009-07-01), pages 3466-3475, XP026104378, ISSN: 0142-9612, DOI: 10.1016/J.BIOMATERIALS.2009.02.040 [retrieved on 2009-03-21]
  • SONG X R ET AL: "Reversion of multidrug resistance by co-encapsulation of vincristine and verapamil in PLGA nanoparticles", EUROPEAN JOURNAL OF PHARMACEUTICAL SCIENCES, ELSEVIER, AMSTERDAM, NL, vol. 37, no. 3-4, 28 June 2009 (2009-06-28), pages 300-305, XP026160257, ISSN: 0928-0987, DOI: 10.1016/J.EJPS.2009.02.018 [retrieved on 2009-03-09]
  • SOMA C E; DUBERNET C; BENTOLILA D; BENITA S; COUVREUR P: "Reversion of multidrug resistance by co-encapsulation of doxorubicin and cyclosporin A in polyalkylcyanoacrylate nanoparticles.", BIOMATERIALS, vol. 21, 1999, - 1999, pages 1-7, XP002699520,
   
Note: Within nine months from the publication of the mention of the grant of the European patent, any person may give notice to the European Patent Office of opposition to the European patent granted. Notice of opposition shall be filed in a written reasoned statement. It shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention).


Description

CROSS-REFERENCE TO RELATED APPLICATIONS



[0001] This application claims priority from U.S. Provisional Application Serial No. 61/333,138 filed on May 10, 2010.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT



[0002] This invention was made with Government support under National Institutes of Health Grant No. U54CA119335 and National Science Foundation Grant No. CMMI-1031239. The Government has certain rights in the invention.

FIELD



[0003] The present teachings relate to nanoparticles, drug conjugates, and controlled release of drug conjugates from the nanoparticles. Methods of making the nanoparticles and drug conjugates are contemplated.

INTRODUCTION



[0004] Combinatorial drug delivery, or combination therapy, refers to the use of multiple drugs to treat diseases or disorders in patients such as various cancers. For example, gemicitabine and paclitaxel are concurrently administered for treating breast cancer; docetaxel and carboplatin for lung cancer; and doxorubicin and ifosfamide for soft tissue sarcoma. Combination chemotherapy is usually more effective than individual chemotherapy as drugs with similar mechanisms act synergistically to enhance therapeutic efficacy whereas drugs with different mechanisms give cancer cells a higher hurdle in developing resistance. However, because of the different therapeutic indices, cellular uptake mechanisms, and in vivo clearance time among drugs, it is difficult to ensure that the tumors receive the optimal dosage of each therapeutic agent. Compositions and methods for precisely controlling the molar ratio among multiple drugs and their concentration taken up by the same target diseased cells would therefore be beneficial in optimizing combination chemotherapy regimens.

[0005] Nanoparticulate drug delivery systems have become increasingly attractive in systemic drug delivery because of their ability to prolong drug circulation half-life, reduce non-specific uptake, and better accumulate at the tumors through enhanced permeation and retention (EPR) effect. As a result, several therapeutic nanoparticles such as Doxil® and Abraxane® are used as the frontline therapies in clinics. But despite the advancement in nanoparticle drug delivery, most research efforts focus on single drug encapsulation. Several strategies have been employed to co-encapsulate multiple drugs into a single nanocarrier, including physical loading into the particle core (see, e.g., X. R. Song, et al. Eur J Pharm Sci 2009, 37, 300-305; C. E. Soma, et al. Biomaterials 2000, 21, 1-7), chemical conjugation to the particle surface (see, e.g., L. Zhang, et al. ChemMedChem 2007, 2, 1268-1271), and covalent linkage to the polymer backbone prior to nanoparticle synthesis (see, e.g., T. Lammers, et al. Biomaterials 2009, 30, 3466-3475; Y. Bae, et al. J Control Release 2007, 122, 324-330; N. Kolishetti, et al. Proc Natl Acad Sci U S A 2010, 107, 17939-17944). However, controlling the ratios of different types of drugs in the same nanoparticles remains a major challenge because of factors such as steric hindrance between the different drug molecules and the polymer backbones, batch-to-batch heterogeneity in conjugation chemistry, and variability in drug-to-drug and drug-to-polymer interactions.

[0006] Many pharmaceutically active agents possess multiple functional groups that are readily modified chemically. Several prodrugs have been synthesized based on these functional groups. For instance, gemcitabine has been acylated through its primary amine to improve its stability in blood; paclitaxel has been pegylated through its hydroxyl groups to improve its water solubility; and doxorubicin has been conjugated to polymers through hydrazone linkage to its ketonic group for nanoparticle encapsulation. It has been demonstrated that modifications through the aforementioned functional groups do not reduce the therapeutic efficacy of chemotherapy drugs as the modified drugs either retain their chemical activities or release the drug content intracellularly through pH- or enzyme-sensitive response.

[0007] Therefore, what is needed are compositions comprising ratiometrically controlled drug combinations, methods of synthesizing such ratiometric compositions, and combination therapy methods of using such compositions.

SUMMARY



[0008] The invention relates to a method for nanoencapsulation of a plurality of drugs comprising:

separately linking each of the plurality of drugs with a corresponding polymer backbone with nearly 100% loading efficiency by forming the corresponding polymer backbone by ring opening polymerization beginning with the corresponding drug, wherein each of the corresponding polymer backbones has the same or similar physicochemical properties and has approximately the same chain length;

mixing the plurality of linked drugs and polymers at selectively predetermined ratios at selectively and precisely controlled drug ratios; and

synthesizing the mixed plurality of linked drugs and polymers into a nanoparticle.



[0009] The present teachings include ratiometric combinatorial drug delivery including nanoparticles, multi-drug conjugates, pharmaceutical compositions and methods of producing such compositions. In one embodiment, a nanoparticle is provided that includes an inner sphere and an outer surface, the inner sphere containing a combination of conjugated drugs connected by a stimuli-sensitive bond and having a predetermined ratio, wherein the conjugated drugs have the following formula:

        (X-Y-Z)n

wherein X is a pharmaceutically active agent, Y is a stimuli-sensitive linker, and Z is not X, and is a pharmaceutically active agent or hydrogen. In various aspects, n is an integer greater than or equal to 2. In another aspect, each individual conjugated drug of the combination comprises a predetermined molar weight percentage from about 1% to about 99%, provided that the sum of all individual conjugated drug molar weight percentages of the combination is 100%. In various aspects of the present embodiment, about 100% of drugs contained in the inner sphere are conjugated.

[0010] In various aspects, X can independently be an antibiotic, antimicrobial, growth factor, chemotherapeutic agent, and combinations thereof. For instance, X can independently include doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof. In various aspects, Z can independently be an antibiotic, antimicrobial, growth factor, chemotherapeutic agent, hydrogen, and combinations thereof. For instance, Z can independently include doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, pharmaceutically acceptable salts thereof, and hydrogen.

[0011] In various aspects, Y is a pH-sensitive linker. For instance, Y can include C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, and combinations thereof.

[0012] In various aspects, the outer surface of the nanoparticle can include a cationic or anionic functional group.

[0013] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula I:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 1 to 10; 'X' is selected from the group consisting of halogen, sulfate, phosphate, nitrate, and water; 'W' is phenyl or tert-butyl oxy; and 'R' is hydrogen or alkyl. For instance, 'p' can be 3; 'X' can be chloride; 'W' can be phenyl and 'R' can be hydrogen.

[0014] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula II:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 1 to 10; 'X' is selected from the group consisting of halogen, sulfate, phosphate, nitrate, and water; 'W1' and 'W2' are independently selected from phenyl or tert-butyl oxy; and 'R' is hydrogen or alkyl. For instance, 'p' can be 3; 'X' is chloride; 'W1' and 'W2' can be phenyl and 'R' can be hydrogen.

[0015] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula III:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 1 to 10; and 'W' is slected from phenyl or tert-butyl oxy. For instance, 'p' can be 3; and 'W' can be phenyl.

[0016] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula IV:

and pharmaceutically acceptable salts thereof, wherein 'W' is phenyl or tert-butyl oxy; and 'V1' and 'V2' are independently selected from -CH3 or -CH2OH. For instance,'W' can be phenyl; and 'V1' and 'V2' can be -CH2OH.

[0017] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula V:

and pharmaceutically acceptable salts thereof, wherein 'W' is phenyl or tert-butyl oxy. For instance, 'W' can be phenyl.

[0018] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula VI:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 5 to 20; and 'W' is phenyl or tert-butyl oxy. For instance, 'p' can be 10; and 'W' can be phenyl.

[0019] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula VII:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 5 to 20; and 'W' is phenyl or tert-butyl oxy. For instance, 'p' can be 10; and 'W' can be phenyl.

[0020] In various aspects, the nanoparticle is about 10 nm to about 10 µm in diameter, and in certain aspects about 30 nm to about 300 nm in diameter.

[0021] In another embodiment, a multi-drug conjugate is provided having the following formula:

        X-Y-Z

wherein X and Z are pharmaceutically active agents independently selected from the group consisting of an antibiotic, antimicrobial, growth factor, and chemotherapeutic agent; and Y is a stimuli-sensitive linker, wherein the conjugate releases at least one pharmaceutically active agent upon delivery of the conjugate to a target cell.

[0022] In various aspects of the present embodiment, Y is a C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, and combinations thereof. For instance, Y can be a C3 straight chain alkyl or a ketone. In various aspects, the pharmaceutically active agent comprises an anticancer chemotherapy agent. For instance, X and Y can independently be doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, or pharmaceutically acceptable salts thereof.

[0023] In yet another aspect, the conjugate has Formula I:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 1 to 10; 'X' is selected from the group consisting of halogen, sulfate, phosphate, nitrate, and water; 'W' is phenyl or tert-butyl oxy; and 'R' is hydrogen or alkyl. For instance, 'p' can be 3; 'X' can be chloride; 'W' can be phenyl and 'R' can be hydrogen.

[0024] In another aspect, the conjugate has Formula II:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 1 to 10; 'X' is selected from the group consisting of halogen, sulfate, phosphate, nitrate, and water; 'W1' and 'W2' are independently selected from phenyl or tert-butyl oxy; and 'R' is hydrogen or alkyl. For instance, 'p' can be 3; 'X' can be chloride; 'W1' and 'W2' can be phenyl and 'R' can be hydrogen.

[0025] In another aspect, the conjugate has Formula III:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 1 to 10; and 'W' is slected from phenyl or tert-butyl oxy. For instance, 'p' can be 3; and 'W' can be phenyl.

[0026] In another aspect, the conjugate has Formula IV:

and pharmaceutically acceptable salts thereof, wherein 'W' is phenyl or tert-butyl oxy; and 'V1' and 'V2' are independently selected from -CH3 or -CH2OH. For instance, 'W' can be phenyl; and 'V1' and 'V2' can be --CH2OH.

[0027] In another aspect, the conjugate has Formula V:

and pharmaceutically acceptable salts thereof, wherein 'W' is phenyl or tert-butyl oxy. For instance, 'W' can be phenyl.

[0028] In another aspect, the conjugate has Formula VI:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 5 to 20; and 'W' is phenyl or tert-butyl oxy. For instance, 'p' can be 10; and 'W' can be phenyl.

[0029] In another aspect, the conjugate has Formula VII:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 5 to 20; and 'W' is phenyl or tert-butyl oxy. For instance, 'p' can be 10; and 'W' can be phenyl.

[0030] In yet another embodiment, a multi-drug conjugate is provided comprising a pharmaceutically active agent covalently bound to a plurality of stimuli-sensitive linkers, wherein each linker is covalently bound to at least one additional pharmaceutically active agent, wherein the conjugate releases at least one pharmaceutically active agent upon delivery to a target cell. In one aspect, the stimuli-sensitive linker can be a C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, or combinations thereof. For instance, the linker can be a C3 straight chain alkyl. In yet another instance, the linker can comprise a ketone.

[0031] In yet another aspect, the pharmaceutically active agent comprises anticancer chemotherapy agents. For instance, the pharmaceutically active agent can include doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.

[0032] In another embodiment, a pharmaceutical composition is provided comprising the multi-drug conjugate above, or a pharmaceutically acceptable salt thereof, in a pharmaceutically acceptable vehicle.

[0033] In yet another embodiment, a method is provided for controlling ratios of conjugated drugs contained in a nanoparticle inner sphere, the method comprising: a) synthesizing a combination of a first drug independently conjugated to a stimuli-sensitive linker, and a second drug independently conjugated to a linker having the same composition, wherein the first drug conjugate and second drug conjugate have a predetermined ratio; b) adding the combination to an agitated solution comprising a polar lipid; and c) adding water to the agitated solution, wherein nanoparticles are produced having a controlled ratio of conjugated drugs contained in the inner sphere. In various aspects of the present embodiment, about 100% of drugs contained in the inner sphere are conjugated.

[0034] In one aspect, the first drug and the second drug can independently include an antibiotic, antimicrobial, antiviral, growth factor, chemotherapeutic agent, and combinations thereof. For instance, the first drug and the second drug are independently selected from the group consisting of doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.

[0035] In another aspect, the stimuli-sensitive linker is a pH-sensitive linker. For instance, the stimuli-sensitive linker is selected from the group consisting of C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, and combinations thereof.

[0036] In various aspects of the present embodiment, the combination of conjugated drugs having a predetermined ratio further comprises at least one additional drug independently conjugated to a stimuli-sensitive linker having the same composition.

[0037] In yet another embodiment, a method is provided for controlling ratios of conjugated drugs contained in a nanoparticle inner sphere, the method comprising: a) synthesizing a combination of (i) a first drug and a second drug conjugated by a first stimuli-sensitive linker, and (ii) a first drug and a second drug conjugated by a second stimuli-sensitive linker, wherein the first drug conjugate and second drug conjugate have a predetermined ratio; b) adding the combination to an agitated solution comprising a polar lipid; and c) adding water to the agitated solution, wherein nanoparticles are produced having a controlled ratio of conjugated drugs contained in the inner sphere. In various aspects of the present embodiment, about 100% of drugs contained in the inner sphere are conjugated.

[0038] In one aspect, the first drug and the second drug are independently selected from the group consisting of an antibiotic, antimicrobial, antiviral, growth factor, chemotherapeutic agent, and combinations thereof. For instance, the first drug and the second drug can independently include doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.

[0039] In another aspect, the stimuli-sensitive linker is a pH-sensitive linker. For instance, the first stimuli-sensitive linker and the second stimuli-sensitive linker can independently include C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, and combinations thereof.

[0040] In various aspects of the present embodiment, the combination of conjugated drugs having a predetermined ratio further comprises at least one additional conjugate of a first drug and a second drug conjugated by a stimuli-sensitive linker other than those present in the combination.

[0041] In another embodiment, a method is provided for producing a combination of conjugated drugs having a predetermined ratio in a nanoparticle, said nanoparticle comprising an inner sphere, the method comprising: a) adding to an agitated solution comprising a polar lipid a combination of a first drug independently conjugated to a stimuli-sensitive linker, and a second drug independently conjugated to a linker having the same composition, wherein the first drug conjugate and the second drug conjugate have a predetermined ratio; and b) adding water to the agitated solution, wherein nanoparticles are produced containing in the inner sphere the conjugated drugs having a predetermined ratio. In various aspects, the method can further comprise: c) isolating nanoparticles having a diameter less than about 300 nm. In various aspects of the present embodiment, about 100% of drugs contained in the inner sphere are conjugated.

[0042] In various aspects, the first drug and the second drug are independently selected from the group consisting of an antibiotic, antimicrobial, growth factor, chemotherapeutic agent, and combinations thereof. For instance, the first drug and the second drug can independently include doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.

[0043] In yet another aspect, the stimuli-sensitive linker is a pH-sensitive linker. For instance, the stimuli-sensitive linker can be C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, or combinations thereof.

[0044] In yet another aspect, the combination of conjugated drugs having a predetermined ratio further comprise a third drug independently conjugated to a stimuli-sensitive linker having the same composition. In various aspects, the solution comprising a polar lipid further comprises a functionlalized polar lipid.

[0045] In yet another embodiment, a method is provided for producing a combination of conjugated drugs having a predetermined ratio in a nanoparticle, said nanoparticle comprising an inner sphere, the method comprising: a) adding to an agitated solution comprising a polar lipid a combination of (i) a first drug and second drug conjugated by a first stimuli-sensitive linker, and (ii) a first drug and a second drug conjugated by a second stimuli-sensitive linker, wherein the first drug conjugate and second drug conjugate have a predetermined ratio; and b) adding water to the agitated solution, wherein nanoparticles are produced containing in the inner sphere the conjugated drugs having a predetermined ratio. In various aspects, the method can further comprise: c) isolating nanoparticles having a diameter less than about 300 nm. In various aspects of the present embodiment, about 100% of drugs contained in the inner sphere are conjugated.

[0046] In one aspect, the first drug and the second drug can independently include an antibiotic, antimicrobial, growth factor, chemotherapeutic agent, and combinations thereof. For instance, the first drug and the second drug are independently selected from the group consisting of doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.

[0047] In another aspect, the stimuli-sensitive linker is a pH-sensitive linker. For instance, the first stimuli-sensitive linker and the second stimuli-sensitive linker can independently be C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, and combinations thereof.

[0048] In various aspects of the present embodiment, the combination of conjugated drugs having a predetermined ratio further comprises at least one additional conjugate of a first drug and a second drug conjugated by a stimuli-sensitive linker other than those present in the combination. In various aspects, the solution comprising a polar lipid further comprises a functionlalized polar lipid.

[0049] In yet another disclosure, a method is provided for treating a disease or condition, the method comprising administering a therapeutically effective amount of the nanoparticle above to a subject in need thereof. In one aspect, the disease is a proliferative disease including lymphoma, renal cell carcinoma, prostate cancer, lung cancer, pancreatic cancer, melanoma, colorectal cancer, ovarian cancer, breast cancer, glioblastoma multiforme and leptomeningeal carcinomatosis. In another aspect, the disease is a heart disease including Atherosclerosis, Ischemic heart disease, Rheumatic heart disease, Hypertensive heart disease, Infective endocarditis, Coronary heart disease, and Constrictive pericarditis. In another aspect, the disease is an ocular disease selected from the group consisting of macular edema, retinal ischemia, macular degeneration, uveitis, blepharitis, keratitis, rubeosis iritis, iridocyclitis, conjunctivitis, and vasculitis. In another aspect, the disease is a lung disease including asthma, Chronic Bronchitis, Cystic Fibrosis, Emphysema, Pneumonia, lung cancer, Primary Pulmonary Hypertension, Pulmonary Arterial Hypertension, and Tuberculosis. In yet another aspect, the disease includes bacterial infection, viral infection, fungal infection, and parasitic infection.

[0050] In various aspects of the present disclosure, the nanoparticle is administered systemically. In another aspect, the nanoparticle is administered locally. In yet another aspect, the local administration is via implantable metronomic infusion pump.

[0051] In yet another disclosure, a method is provided for treating a disease or condition, the method comprising administering a therapeutically effective amount of the multi-drug conjugate above to a subject in need thereof. In one aspect, the disease is a proliferative disease including lymphoma, renal cell carcinoma, prostate cancer, lung cancer, pancreatic cancer, melanoma, colorectal cancer, ovarian cancer, breast cancer, glioblastoma multiforme and leptomeningeal carcinomatosis. In one aspect, the disease is a heart disease including Atherosclerosis, Ischemic heart disease, Rheumatic heart disease, Hypertensive heart disease, Infective endocarditis, Coronary heart disease, and Constrictive pericarditis. In one aspect, the disease is an ocular disease including macular edema, retinal ischemia, macular degeneration, uveitis, blepharitis, keratitis, rubeosis iritis, iridocyclitis, conjunctivitis, and vasculitis. In one aspect, the disease is a lung disease including asthma, Chronic Bronchitis, Cystic Fibrosis, Emphysema, Pneumonia, lung cancer, Primary Pulmonary Hypertension, Pulmonary Arterial Hypertension, and Tuberculosis. In yet another aspect, the disease is selected from the group consisting of bacterial infection, viral infection, fungal infection, and parasitic infection.

[0052] In various aspects of the present disclosure, the multi-drug conjugate is administered systemically. In another aspect, the multi-drug conjugate is administered locally. In yet another aspect, the local administration is via implantable metronomic infusion pump.

[0053] In yet another disclosure, a method is provided for sequentially delivering a drug conjugate to a target cell, the method comprising administering a nanoparticle above to the target cell and triggering multi-drug conjugate release. In various aspects of the present embodiment, the nanoparticle is administered systemically. In another aspect, the nanoparticle is administered locally. In yet another aspect, the local administration is via implantable metronomic infusion pump.

[0054] In yet another embodiment, a method is provided for nanoencapsulation of a plurality of drugs comprising separately linking each of the plurality of drugs with a corresponding polymer backbone with nearly 100% loading efficiency by forming the corresponding polymer backbone by ring opening polymerization beginning with the corresponding drug, wherein each of the corresponding polymer backbones has the same or similar physicochemical properties and has approximately the same chain length; mixing the plurality of linked drugs and polymers at selectively predetermined ratios at selectively and precisely controlled drug ratios; and synthesizing the mixed plurality of linked drugs and polymers into a nanoparticle.

[0055] In various aspects, the plurality of drugs can independently include an antibiotic, antimicrobial, growth factor, chemotherapeutic agent, and combinations thereof. For instance, the plurality of drugs can independently include doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.

[0056] In various aspects, the polymer backbone is a stimuli-sensitive linker. For instance, the stimuli-sensitive linker can include a C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, and combinations thereof.

[0057] These and other features, aspects and advantages of the present teachings will become better understood with reference to the following description, examples and appended claims.

DRAWINGS



[0058] Those of skill in the art will understand that the drawings, described below, are for illustrative purposes only. The drawings are not intended to limit the scope of the present teachings in any way.

Figure 1. Schematic illustration of a dual-drug loaded lipid-polymer hybrid nanoparticle, of which the polymeric core consists of two distinct drug-polymer conjugates with ratiometric control over drug loading.

Figure 2. Chemical characterization of the drug-polymer conjugates. (A) Schematic description of the living ring-opening polymerization of 1-lactide catalyzed by an activated metal alkoxide complex. (B) Qualitative 1H-NMR spectra showing the characteristic proton resonance peaks of DOX-PLA (upper panel) and CPT-PLA (lower panel). (C) Gel permeation chromatograms of DOX-PLA (red dashed line) and CPT-PLA (black solid line).

Figure 3. Scanning electron microscopy (SEM) and dynamic light scattering (DLS) measurements showing the morphology and size of lipid-polymer hybrid nanoparticles with the polymer cores consisting of: (A) DOX-PLA conjugates, (B) CPT-PLA conjugates, or (C) DOX-PLA and CPT-PLA conjugates with a molar ratio of 1:1.

Figure 4. Quantification of DOX and CPT loading efficiency in dual-drug loaded nanoparticles (containing both DOX-PLA and CAP-PLA) and single-drug loaded nanoparticles (containing DOX-PLA or CPT-PLA), respectively. NPs: nanoparticles.

Figure 5. Cellular colocalization and cytotoxicity studies of the DOX-PLA and CPT-PLA loaded dual-drug nanoparticles. (A) Fluorescence microscopy images showing the colocalization of DOX and CPT in the cellular compartment of MDB-MB-435 breast cancer cells. (B) A comparative study of cellular cytotoxicity of the DOX-PLA and CPT-PLA loaded dual-drug nanoparticles against the MDB-MB-435 breast cancer cells. The ratios shown in figure legends are the molar ratios of DOX-PLA to CPT-PLA. Solid lines represent the dual-drug loaded nanoparticles and dashed lines represent the cocktail mixture of DOX-PLA loaded and CPT-PLA loaded single-drug nanoparticles. All samples were incubated with cells for 24 h, and the cells were subsequently washed and incubated in media for a total of 72 h prior to MTT assay (n=4).

Figure 6. Mass spectrum (ESI-positive ion mode) of 2-((2,6-diisopropylphenyl)amido)-4-((2,6diisopropylphenyl)-imino)-2-pentene (BDI).

Figure 7. 1H-NMR characterization of 2-((2,6-diisopropylphenyl)amido)-4-((2,6diisopropylphenyl) -imino)-2-pentene (BDI).

Figure 8. 1H-NMR characterization of (BDI)ZnN(SiMe3)2 complex catalyst.

Figure 9. Synthesis scheme of paclitaxel (PTXL) and gemcitabine hydrochloride (GEM) conjugate (PTXL-GEM conjugate, compound 2).

Figure 10. Characterization of PTXL-GEM conjugates using (A) 1H-NMR spectroscopy showing the characteristic protons, and (B) high resolution mass spectrum determining the exact mass and corresponding molecular formula of the drug conjugates.

Figure 11. Hydrolysis and cellular cytotoxicity of PTXL-GEM conjugates. (A) HPLC chromatograms of PTXL-GEM conjugates (a) before and (b) after 24 hrs of incubation in water/acetonitrile (75/25, v/v) solution at pH = 7.4. (B) Hydrolysis kinetics of PTXL-GEM conjugates at pH = 6.0 and pH = 7.4. (C) Time dependent comparative cytotoxicity of PTXL-GEM conjugates with the corresponding mixture of free PTXL and free GEM drugs at 100 nM concentration against XPA3 human pancreatic cancer cell line (n = 8).

Figure 12. Characterization of PTXL-GEM conjugates loaded lipid-coated polymeric nanoparticles (NPs). (A) Schematic illustration of a PTXL-GEM conjugates loaded nanoparticle. (B) Representative scanning electron microscopy (SEM) image of PTXL-GEM conjugates loaded nanoparticles. (C) Diameter and surface zeta-potential of PTXL-GEM conjugates loaded nanoparticles and empty nanoparticles measured by dyanamic light scattering (DLS).

Figure 13. (A) PTXL-GEM conjugates loading yield at various initial weight ratios of PTXL-GEM conjugates/excipient (PLGA polymer). (B) Cellular cytotoxicity of PTXL-GEM conjugates loaded nanoparticles and free PTXL-GEM conjugates (compound 2) at various drug conjugate concentrations against XPA3 human pancreatic cancer cell line. All samples were incubated with cells for 24 hrs, and the cells were subsequently washed and incubated in media for a total of 72 hrs before assessing cell viability in each group (n = 8).

Figure 14. 1H NMR spectrum of paclitaxel.

Figure 15. 1H NMR spectrum of compound 1.

Figure 16. ESI-MS (positive) mass spectrum of compound 1.

Figure 17. ESI-MS (positive) mass spectrum of paclitaxel recovered from the hydrolyzed PTXL-GEM conjugates with an HPLC retention time of 6.2 min.

Figure 18. ESI-MS (positive) mass spectrum of gemcitabine recovered from the hydrolyzed PTXL-GEM conjugates with an HPLC retention time of 1.8 min.

Figure 19. Synthesis scheme of paclitaxel (Ptxl) and cisplatin conjugate (Ptxl-Pt(IV) conjugate) as a representative hydrophobic-hydrophilic drug conjugate.

Figure 20. Characterization of Ptxl-Pt(IV) conjugate using (A) 1H-NMR spectroscopy showing the characteristic protons, and (B) high resolution mass spectrum determining the exact mass and corresponding molecular formula of the Ptxl-Pt(IV) conjugate.

Figure 21. Characterization of Ptxl-Pt(IV) conjugates loaded nanoparticles. (A) Schematic illustration of Ptxl-Pt(IV) conjugates loaded lipid coated polymeric nanoparticles. (B) Dynamic light scattering (DLS) measurement of Ptxl-Pt(IV) loaded nanoparticles. (C) Representative scanning electron microscopy (SEM) image of Ptxl-Pt(IV) loaded nanoparticles. Inset: high-resolution SEM image of Ptxl-Pt(IV) loaded nanoparticles

Figure 22. (A) Cellular cytotoxicity of free Ptxl-Pt(IV) conjugates and Ptxl-Pt(IV) conjugates loaded nanoparticles (NPs) at various drug concentration against A2780 human ovarian cancer cell line. All samples were incubated with cells for 24 hrs, and the cells were subsequently washed and incubated in fresh media for a total of 72 hrs before cell viability using the ATP assay (n=8). (B,C) Representative phase contrast microscopy images of A2780 cells treated with (B) free Ptxl-Pt(IV) drug conjugates and (C) Ptxl-Pt(IV) conjugates loaded nanoparticles, respectively, at a drug concentration of 300 nM.

Figure 23. 1H NMR spectrum of cis-trans-cis PtCl2(OCOCH2CH2CH2COOH)2(NH3)2.

Figure 24. Drug loading yield of PTXL conjugates.


DETAILED DESCRIPTION


Abbreviations and Definitions



[0059] To facilitate understanding of the invention, a number of terms and abbreviations as used herein are defined below as follows:

[0060] When introducing elements of the present invention or the preferred embodiment(s) thereof, the articles "a", "an", "the" and "said" are intended to mean that there are one or more of the elements. The terms "comprising", "including" and "having" are intended to be inclusive and mean that there may be additional elements other than the listed elements.

[0061] The term "and/or" when used in a list of two or more items, means that any one of the listed items can be employed by itself or in combination with any one or more of the listed items. For example, the expression "A and/or B" is intended to mean either or both of A and B, i.e. A alone, B alone or A and B in combination. The expression "A, B and/or C" is intended to mean A alone, B alone, C alone, A and B in combination, A and C in combination, B and C in combination or A, B, and C in combination.

[0062] In the descriptions of molecules and substituents, molecular descriptors can be combined to produce words or phrases that describe substituents. Such descriptors are used in this document. Examples include such terms as aralkyl (or arylalkyl), heteroaralkyl, heterocycloalkyl, cycloalkylalkyl, aralkoxyalkoxycarbonyl and the like. A specific example of a compound encompassed with the latter descriptor aralkoxyalkoxycarbonyl is C6H5-CH2-CH2-O-CH2-O-C(O) wherein C6H5 is phenyl. It is also to be noted that a substituents can have more than one descriptive word or phrase in the art, for example, heteroaryloxyalkylcarbonyl can also be termed heteroaryloxyalkanoyl. Such combinations are used herein in the description of the compounds and methods of this invention and further examples are described herein.

[0063] Alkyl: The term "alkyl" as used herein describes substituents which are preferably lower alkyl containing from one to eight carbon atoms in the principal chain and up to about 20 carbon atoms. The principal chain may be straight or branched chain or cyclic and include methyl, ethyl, propyl, isopropyl, butyl, hexyl and the like.

[0064] Analog: The term "analog" as used herein may refer to a compound in which one or more atoms are replaced with a different atom or group of atoms. The term may also refer to compounds with an identity of atoms but of different isomeric configuration. Such isomers may be constitutional isomers, i.e. structural isomers having different bonding arrangements of their atoms or stereoisomers having identical bonding arrangements but different spatial arrangements of the constituent atoms.

[0065] Anionic: The term "anionic" as used herein refers to substances capable of forming ions in aqueous media with a net negative charge.

[0066] Anionic functional group: The term "anionic functional group" as used herein refers to functional group as defined herein which possesses a net negative charge. Representative anionic functional groups include carboxylic, sulfonic, phosphonic, their alkylated derivatives, and so on.

[0067] Cationic: The term "cationic" as used herein refers to substances capable of forming ions in aqueous media with a net positive charge.

[0068] Functional group: The term "functional group" as used herein, refers to a chemical group that imparts a particular function to an article (e.g., nanoparticle) bearing the chemical group. For example, functional groups can include substances such as antibodies, oligonucleotides, biotin, or streptavidin that are known to bind particular molecules; or small chemical groups such as amines, carboxylates, and the like.

[0069] Halogen: The terms "halogen" or "halo" as used herein, alone or as part of a group of atoms, refer to chlorine, bromine, fluorine, and iodine.

[0070] Nanoparticle: The term "nanoparticle" as used herein refers to unilamellar or multilamellar lipid vesicles which enclose a fluid space and has a diameter of between about 1 nm and about 1000 nm. Similarly, by the term "nanoparticles" is meant a plurality of particles having an average diameter of between about 1 nm and about 1000 nm. The term can also include vesicles as large as 10,000 nm depending on the environment such nanoparticles are administered to a subject, for example, locally to a tumor in situ via implantable pump or via syringe. For systemic use, an average diameter of about 30 nm to about 300 nm is preferred. The walls of the vesicles, also referred to as a membrane, are formed by a bimolecular layer of one or more lipid components (e.g., multiple phospholipids and cholesterol) having polar heads and non-polar tails, such as a phospholipid. In an aqueous (or polar) solution, and in a unilamellar nanoparticle, the polar heads of one layer orient outwardly to extend into the surrounding medium, and the non-polar tail portions of the lipids associate with each other, thus providing a polar surface and a non-polar core in the wall of the vesicle. In a multilamellar nanoparticle, the polar surface of the vesicle also extends to the core of the liposome and the wall is a bilayer. The wall of the vesicle in either of the unilamellar or multilamellar nanoparticles can be saturated or unsaturated with other lipid components, such as cholesterol, free fatty acids, and phospholipids. In such cases, an excess amount of the other lipid component can be added to the vesicle wall which will shed until the concentration in the vesicle wall reaches equilibrium, which can be dependent upon the nanoparticle environment. Nanoparticles may also comprise other agents that may or may not increase an activity of the nanoparticle. For example, polyethylene glycol (PEG) can be added to the outer surface of the membrane to enhance bioavailability. In other examples, functional groups such as antibodies and aptamers can be added to the outer surface of the membrane to enhance site targeting, such as to cell surface epitopes found in cancer cells. The membrane of the nanoparticles can also comprise particles that can be biodegradable, cationic nanoparticles including, but not limited to, gold, silver, and synthetic nanoparticles. An example of a biocompatible synthetic nanoparticle includes polystyrene and the like.

[0071] Pharmaceutically active: The terms "pharmaceutically active" as used herein refer to the beneficial biological activity of a substance on living matter and, in particular, on cells and tissues of the human body. A "pharmaceutically active agent" or "drug" is a substance that is pharmaceutically active and a "pharmaceutically active ingredient" (API) is the pharmaceutically active substance in a drug.

[0072] Pharmaceutically acceptable: The terms "pharmaceutically acceptable" as used herein means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopoeia, other generally recognized pharmacopoeia in addition to other formulations that are safe for use in animals, and more particularly in humans and/or non-human mammals.

[0073] Pharmaceutically acceptable salt: The terms "pharmaceutically acceptable salt" as used herein refer to acid addition salts or base addition salts of the compounds, such as the multi-drug conjugates, in the present disclosure. A pharmaceutically acceptable salt is any salt which retains the activity of the parent compound and does not impart any deleterious or undesirable effect on a subject to whom it is administered and in the context in which it is administered. Pharmaceutically acceptable salts include, but are not limited to, metal complexes and salts of both inorganic and carboxylic acids. Pharmaceutically acceptable salts also include metal salts such as aluminum, calcium, iron, magnesium, manganese and complex salts. In addition, pharmaceutically acceptable salts include, but are not limited to, acid salts such as acetic, aspartic, alkylsulfonic, arylsulfonic, axetil, benzenesulfonic, benzoic, bicarbonic, bisulfuric, bitartaric, butyric, calcium edetate, camsylic, carbonic, chlorobenzoic, citric, edetic, edisylic, estolic, esyl, esylic, formic, fumaric, gluceptic, gluconic, glutamic, glycolic, glycolylarsanilic, hexamic, hexylresorcjnoic, hydrabamic, hydrobromic, hydrochloric, hydroiodic, hydroxynaphthoic, isethionic, lactic, lactobionic, maleic, malic, malonic, mandelic, methanesulfonic, methylnitric, methylsulfuric, mucic, muconic, napsylic, nitric, oxalic, p-nitromethanesulfonic, pamoic, pantothenic, phosphoric, monohydrogen phosphoric, dihydrogen phosphoric, phthalic, polygalactouronic, propionic, salicylic, stearic, succinic, sulfamic, sulfanlic, sulfonic, sulfuric, tannic, tartaric, teoclic, toluenesulfonic, and the like. Pharmaceutically acceptable salts may be derived from amino acids including, but not limited to, cysteine. Methods for producing compounds as salts are known to those of skill in the art (see, for example, Stahl et al., Handbook of Pharmaceutical Salts: Properties, Selection, and Use, Wiley-VCH; Verlag Helvetica Chimica Acta, Zürich, 2002; Berge et al., J Pharm. Sci. 66: 1, 1977).

[0074] Pharmaceutically acceptable carrier: The terms "pharmaceutically acceptable carrier" as used herein refers to an excipient, diluent, preservative, solubilizer, emulsifier, adjuvant, and/or vehicle with which a compound, such as a multi-drug conjugate, is administered. Such carriers may be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents. Water is a preferred carrier when a compound is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions may also be employed as liquid carriers, particularly for injectable solutions. Suitable excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. A compound, if desired, may also combine minor amounts of wetting or emulsifying agents, or pH buffering agents such as acetates, citrates or phosphates. Antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; and agents for the adjustment of tonicity such as sodium chloride or dextrose may also be a carrier. Methods for producing compounds in combination with carriers are known to those of skill in the art.

[0075] Phospholipid: The term "phospholipid", as used herein, refers to any of numerous lipids contain a diglyceride, a phosphate group, and a simple organic molecule such as choline. Examples of phospholipids include, but are not limited to, Phosphatidic acid (phosphatidate) (PA), Phosphatidylethanolamine (cephalin) (PE), Phosphatidylcholine (lecithin) (PC), Phosphatidylserine (PS), and Phosphoinositides which include, but are not limited to, Phosphatidylinositol (PI), Phosphatidylinositol phosphate (PIP), Phosphatidylinositol bisphosphate (PIP2) and Phosphatidylinositol triphosphate (PIP3). Additional examples of PC include DDPC, DLPC, DMPC, DPPC, DSPC, DOPC, POPC, DRPC, and DEPC as defined in the art.

[0076] Stimuli-Sensitive Linker: As used herein, the term "stimuli-sensitive linker" refers to a carbon chain that can contain heteroatoms (e.g., nitrogen, oxygen, sulfur, etc.) and which may be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50 atoms long. Stimuli-sensitive linkers may be substituted with various substituents including, but not limited to, hydrogen atoms, alkyl, alkenyl, alkynl, amino, alkylamino, dialkylamino, trialkylamino, hydroxyl, alkoxy, halogen, aryl, heterocyclic, aromatic heterocyclic, cyano, amide, carbamoyl, carboxylic acid, ester, thioether, alkylthioether, thiol, and ureido groups. Those of skill in the art will recognize that each of these groups may in turn be substituted. Examples of stimuli-sensitive linkers include, but are not limited to, pH sensitive linkers, protease cleavable peptide linkers, nuclease sensitive nucleic acid linkers, lipase sensitive lipid linkers, glycosidase sensitive carbohydrate linkers, hypoxia sensitive linkers, photo-cleavable linkers, heat-labile linkers, enzyme cleavable linkers (e.g., esterase cleavable linker), ultrasound-sensitive linkers, x-ray cleavable linkers, and so forth.

[0077] Substituted: The term "substituted" as used herein refers to one or more substitutions that are common in the art. The terms "optionally substituted" means that a group may be unsubstituted or substituted with one or more substituents. Suitable substituents for any of the groups defined above may include moieties such as alkyl, cycloalkyl, alkenyl, alkylidenyl, aryl, heteroaryl, heterocyclyl, halo (e.g., chloro, bromo, iodo and fluoro), cyano, hydroxy, alkoxyl, aroxyl, sulfhydryl (mercapto), alkylthio, arylthio, amino, substituted amino, nitro, carbamyl, keto (oxo), acyl, glycolyl, glycyl, hydrazino, guanyl, sulfamyl, sulfonyl, sulfinyl, thioalkyl-C(O)--, thioalkyl-CO2--, and the like.

[0078] Therapeutically Effective Amount: As used herein, the term "therapeutically effective amount" refers to those amounts that, when administered to a particular subject in view of the nature and severity of that subject's disease or condition, will have a desired therapeutic effect, e.g., an amount which will cure, prevent, inhibit, or at least partially arrest or partially prevent a target disease or condition. More specific embodiments are included in the Pharmaceutical Preparations and Methods of Administration section below.

Ratiometric Combinatorial Drug Delivery



[0079] The present teachings include ratiometric combinatorial drug delivery including nanoparticles, multi-drug conjugates, pharmaceutical compositions, methods of producing such compositions and methods of using such compositions, including in the treatment of diseases and conditions using drug combinations.

[0080] A combinatorial drug conjugation approach is provided to enable multi-drug delivery. In one example, hydrophobic and hydrophilic drugs were covalently conjugated using a hydrolysable linker and then encapsulated into lipid-polymer hybrid nanoparticles for combined delivery. In one non-limiting example, the ratio between two drugs co-delivered, some with drastically different properties, included various ratios including a 1:1 drug-drug ratio, and in other examples 3:1 and 1:3 ratios. As disclosed herein, such ratios can be controlled by the different molar amounts of the drugs in combination which results in versatile multi-drug encapsulation schemes.

[0081] In one aspect, each different drug molecule is linked to an individual linker backbone that has the same physicochemical properties and nearly the same chain length (i.e. a drug-linker). These drug-linker conjugates can be subsequently mixed at predetermined ratios prior to or during nanoparticle synthesis. The long and sharply distributed linker, in some examples a polymer chain, can provide each drug molecule a predominant and uniform hydrophobic property, and yield near 100% drug loading efficiency upon nanoparticle formation. In various aspects, the linkers can be stimuli-sensitive such that the linked drug is cleaved upon a change in the nanoparticle or multi-drug conjugate environment, such as a difference in pH.

[0082] In another aspect, an individual drug molecule is linked to another individual drug molecule, each being linked through different linkers. These drug-drug conjugates can be subsequently mixed or created at predetermined ratios prior to or during nanoparticle synthesis. The hydrophobic properties of these conjugates can be different and the linkers can have different stimuli-sensitive activities. This can result in sequential drug delivery as one linker can be cleaved to release a drug at a certain environmental state, and a second linker can release the same or different drug upon a change in environmental state, such as a different pH.

[0083] As provided in one non-limiting example, the synthesis of a drug-linker conjugate with two different pharmaceutically active agents, doxorubicin (DOX) and camptothecin (CPT), is provided. Utilizing ring-opening polymerization of 1-lactide, DOX and CPT polymer conjugates were synthesized using metal-amido catalyst, which reacts selectively with hydroxyl groups of the drug molecules to initiate polymerization (R. Tong, J. Cheng, Angew Chem Int Ed Engl 2008, 47, 4830-4834; R. Tong, J. Cheng, Angew Chem 2008, 120, 4908-4912; R. Tong, J. Cheng, Bioconjug Chem 2010, 21, 111-121; R. Tong, J. Cheng, J Am Chem Soc 2009, 131, 4744-4754). Using a nanoprecipitation technique (Figure 1), the drug-polymer conjugates were quantitatively loaded into lipid-polymer hybrid nanoparticles at high loading efficiency and precisely controlled drug ratios. See B. M. Chamberlain, et al. J Am Chem Soc 2001, 123, 3229-3238; L. Zhang, et al. ACS Nano 2008, 2, 1696-1702. The combinatorial treatment provided herein shows superior efficacy to cocktail therapy in vitro and offers a solution to the aforementioned limitations in multi-drug encapsulation into the same nanoparticles.

Ratiometrically Controlled Nanoparticles



[0084] Therefore, in one embodiment, a nanoparticle is provided that includes an inner sphere and an outer surface, the inner sphere containing a combination of conjugated drugs connected by a stimuli-sensitive bond and having a predetermined ratio, wherein the conjugated drugs have the following formula:

        (X-Y-Z)n

wherein X is a pharmaceutically active agent, Y is a stimuli-sensitive linker, and Z is not X, and Z is a pharmaceutically active agent or hydrogen.

[0085] In various aspects, X and Z can independently be an antibiotic, antimicrobial, growth factor, chemotherapeutic agent, and combinations thereof. A listing of classes and specific drugs suitable for use in the present invention may be found in Pharmaceutical Drugs: Syntheses, Patents, Applications by Axel Kleemann and Jurgen Engel, Thieme Medical Publishing, 1999 and the Merck Index: An Encyclopedia of Chemicals, Drugs and Biologicals, Ed. by Budavari et al., CRC Press, 1996, both of which are incorporated herein by reference. Examples of such pharmaceutically active agents are provided in the Tables appended hereto. Such pharmaceutically active agents can be delivered to particular organs, tissues, cells, extracellular matrix components, and/or intracellular compartments via any suitable method, including the use of a functional group such as an antibody, antibody fragment, aptamer, and so on.

[0086] For instance, X can independently include doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.

[0087] These and other pharmaceutically active agents can be covalently conjugated by a suitable chemical linker through environmentally cleavable bonds. Any of a variety of methods can be used to associate a linker with a pharmaceutically active agent including, but not limited to, passive adsorption (e.g., via electrostatic interactions), multivalent chelation, high affinity non-covalent binding between members of a specific binding pair, covalent bond formation, etc. In some embodiments, click chemistry can be used to associate a linker with a particle (e.g. Diels-Alder reaction, Huigsen 1,3-dipolar cycloaddition, nucleophilic substitution, carbonyl chemistry, epoxidation, dihydroxylation, etc.). In various aspects, drug conjugates including a plurality of pharmaceutically active agents, each of which is covalently bound to a linker, wherein the conjugate releases the pharmaceutically active agent upon delivery to target cells, are provided.

[0088] Some chemical bonds such as hydrazone, ester and amide bonds are sensitive to acidic pH values, for example, of the intracellular environment of tumor cells. At acidic pH, hydrogen ions catalyze the hydrolysis of these bonds which in turn releases the drug from its conjugate format. Therefore, different pharmaceutically active agents, such as but not limited to paclitaxel, gemcitabin, doxorubicine, cisplatin, docetaxel, etc, having -OH, -NH2, and/or ketonic groups may be covalently linked together with a suitable spacer with alkyl chains of variable lengths. These spacers may be easily introduced to the drug conjugates by reacting different acid anhydrides and any organic compounds having monofunctional or bifunctional or hetero functional groups with the drugs.

[0089] For the pharmaceutically active agents without functional groups such as-OH, -NH2, or ketonic groups, they may be covalently linked with other pharmaceutically active agents by creating such functional groups. For example, cisplatin can first be oxidized to its hydroxyl derivative which then can react with carboxylic acid aldehyde or acid anhydride to create an aldehydic and carboxylic functional group. This functional group can be covalently linked with other drugs with -OH and/or -NH2. Many pharmaceutically active agents can be linked together to form combinatorial drug conjugates for combination therapy. Those of skill in the art are able to recognize other conjugation methods which are well known in the art. Such conjugation methods may be used to link various pharmaceutically active agents, including small molecules, polypeptides, and polynucleotides, via linkers, including stimuli-sensitive linkers.

[0090] In various aspects, the variable 'n' of the formula (X-Y-Z)n is an integer greater than or equal to 2. In various aspects, this numeral represents 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50 and even greater numbers of drug-linker and drug-drug conjugates can be contained in the nanoparticle.

[0091] In another aspect, each individual conjugated drug of the combination comprises a predetermined molar weight percentage from about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, to about 99%, provided that the sum of all individual conjugated drug molar weight percentages of the combination is 100%. For example, a first drug-linker conjugate can comprise 70 weight percent (70% w/w) and a second drug-linker conjugate can comprise 30 weight percent (30% w/w) as contained in the nanoparticle. In another example, a first drug-drug conjugate can comprise 40 weight percent (40% w/w) and a second drug-linker conjugate can comprise 60 weight percent (60% w/w) as contained in the nanoparticle. In yet another example, a first drug-linker conjugate can comprise 10 weight percent (10% w/w), a second drug-linker conjugate can comprise 30 weight percent (30% w/w), and a third drug-linker conjugate can comprise 60 weight percent (60% w/w) as contained in the nanoparticle. As another example, a first drug-drug conjugate can comprise 10 weight percent (10% w/w), a second drug-drug conjugate can comprise 30 weight percent (30% w/w), and a third drug-drug conjugate can comprise 60 weight percent (60% w/w) as contained in the nanoparticle.

[0092] By using predetermined molar weight percentages, precise ratios among conjugated drugs in the nanoparticle can be provided. For example, among two-drug conjugate combinations, ratios including 1:1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, 327, 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 340, 341, 342, 343, 344, 345, 346, 347, 348, 349, 350, 351, 352, 353, 354, 355, 356, 357, 358, 359, 360, 361, 362, 363, 364, 365, 366, 367, 368, 369, 370, 371, 372, 373, 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384, 385, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396, 397, 398, 399, 400, 401, 402, 403, 404, 405, 406, 407, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, 431, 432, 433, 434, 435, 436, 437, 438, 439, 440, 441, 442, 443, 444, 445, 446, 447, 448, 449, 450, 451, 452, 453, 454, 455, 456, 457, 458, 459, 460, 461, 462, 463, 464, 465, 466, 467, 468, 469, 470, 471, 472, 473, 474, 475, 476, 477, 478, 479, 480, 481, 482, 483, 484, 485, 486, 487, 488, 489, 490, 491, 492, 493, 494, 495, 496, 497, 498, 499, and 1:500 are provided. In another example having three-drug conjugate combinations ratios of 1:1:1, 1:2:1, 1:3:1, 1:1:2, 1:1:3, and so forth are provided. Those of skill in the art will recognize that other ratios can be provided with different numbers of drugs and different molar weight percentages are utilized.

[0093] In various aspects, Z can independently be an antibiotic, antimicrobial, growth factor, chemotherapeutic agent, hydrogen, and combinations described above. In addition, Z can be hydrogen (e.g., a drug-linker conjugate).

[0094] In various aspects, Y is a pH-sensitive linker. For instance, Y can include C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, and combinations thereof.

[0095] In various aspects, the outer surface of the nanoparticle can include a cationic or anionic functional group.

[0096] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula I:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 1 to 10; 'X' is selected from the group consisting of halogen, sulfate, phosphate, nitrate, and water; 'W' is phenyl or tert-butyl oxy; and 'R' is hydrogen or alkyl. For instance, 'p' can be 3; 'X' can be chloride; 'W' can be phenyl and 'R' can be hydrogen.

[0097] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula II:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 1 to 10; 'X' is selected from the group consisting of halogen, sulfate, phosphate, nitrate, and water; 'W1' and 'W2' are independently selected from phenyl or tert-butyl oxy; and 'R' is hydrogen or alkyl. For instance, 'p' can be 3; 'X' is chloride; 'W1' and 'W2' can be phenyl and 'R' can be hydrogen.

[0098] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula III:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 1 to 10; and 'W' is slected from phenyl or tert-butyl oxy. For instance, 'p' can be 3; and 'W' can be phenyl.

[0099] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula IV:

and pharmaceutically acceptable salts thereof, wherein 'W' is phenyl or tert-butyl oxy; and 'V1' and 'V2' are independently selected from -CH3 or -CH2OH. For instance, 'W' can be phenyl; and 'V1' and 'V2' can be -CH2OH.

[0100] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula V:

and pharmaceutically acceptable salts thereof, wherein 'W' is phenyl or tert-butyl oxy. For instance, 'W' can be phenyl.

[0101] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula VI:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 5 to 20; and'W' is phenyl or tert-butyl oxy. For instance, 'p' can be 10; and 'W' can be phenyl.

[0102] In yet another aspect, the conjugated drug of the combination contained in the nanoparticle inner sphere has Formula VII:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 5 to 20; and 'W' is phenyl or tert-butyl oxy. For instance, 'p' can be 10; and 'W' can be phenyl.

[0103] In various aspects, the nanoparticle can be about 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, 327, 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 340, 341, 342, 343, 344, 345, 346, 347, 348, 349, 350, 351, 352, 353, 354, 355, 356, 357, 358, 359, 360, 361, 362, 363, 364, 365, 366, 367, 368, 369, 370, 371, 372, 373, 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384, 385, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396, 397, 398, 399, 400, 401, 402, 403, 404, 405, 406, 407, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, 431, 432, 433, 434, 435, 436, 437, 438, 439, 440, 441, 442, 443, 444, 445, 446, 447, 448, 449, 450, 451, 452, 453, 454, 455, 456, 457, 458, 459, 460, 461, 462, 463, 464, 465, 466, 467, 468, 469, 470, 471, 472, 473, 474, 475, 476, 477, 478, 479, 480, 481, 482, 483, 484, 485, 486, 487, 488, 489, 490, 491, 492, 493, 494, 495, 496, 497, 498, 499, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, and about 10000 nm in diameter. In various aspects, and particularly depending on the route of administration in a subject, the nanoparticle can have a diameter from about 30 nm to about 300 nm. In general, larger nanoparticles are acceptable when administered locally or topically where the nanoparticle is not required to traverse a subject vasculature to contact a target cell, tissue or organ. Likewise, smaller nanoparticles are acceptable when administered systemically in a subject, in particular nanoparticles from about 30 nm to about 300 nm.

Multi-Drug Conjugates



[0104] In another embodiment, a multi-drug conjugate is provided having the following formula:

        X-Y-Z

wherein X and Z are pharmaceutically active agents independently selected from the group consisting of an antibiotic, antimicrobial, growth factor, and chemotherapeutic agent; and Y is a stimuli-sensitive linker, wherein the conjugate releases at least one pharmaceutically active agent upon delivery of the conjugate to a target cell. Such conjugated drugs are provided above as contained in the nanoparticle of the present invention.

[0105] In various aspects of the present embodiment, Y is a C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, and combinations thereof. For instance, Y can be a C3 straight chain alkyl or a ketone. In various aspects, the pharmaceutically active agent comprises an anticancer chemotherapy agent. For instance, X and Y can independently be doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, or pharmaceutically acceptable salts thereof.

[0106] In yet another aspect, the conjugate has Formula I:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 1 to 10; 'X' is selected from the group consisting of halogen, sulfate, phosphate, nitrate, and water; 'W' is phenyl or tert-butyl oxy; and 'R' is hydrogen or alkyl. For instance, 'p' can be 3; 'X' can be chloride; 'W' can be phenyl and 'R' can be hydrogen.

[0107] In another aspect, the conjugate has Formula II:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 1 to 10; 'X' is selected from the group consisting of halogen, sulfate, phosphate, nitrate, and water; 'W1' and 'W2' are independently selected from phenyl or tert-butyl oxy; and 'R' is hydrogen or alkyl. For instance, 'p' can be 3; 'X' can be chloride; 'W1' and 'W2' can be phenyl and 'R' can be hydrogen.

[0108] In another aspect, the conjugate has Formula III:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 1 to 10; and 'W' is slected from phenyl or tert-butyl oxy. For instance, 'p' can be 3; and 'W' can be phenyl.

[0109] In another aspect, the conjugate has Formula IV:

and pharmaceutically acceptable salts thereof, wherein 'W' is phenyl or tert-butyl oxy; and 'V1' and 'V2' are independently selected from -CH3 or -CH2OH. For instance, 'W' can be phenyl; and 'V1' and 'V2' can be --CH2OH.

[0110] In another aspect, the conjugate has Formula V:

and pharmaceutically acceptable salts thereof, wherein 'W' is phenyl or tert-butyl oxy. For instance, 'W' can be phenyl.

[0111] In another aspect, the conjugate has Formula VI:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 5 to 20; and 'W' is phenyl or tert-butyl oxy. For instance, 'p' can be 10; and 'W' can be phenyl.

[0112] In another aspect, the conjugate has Formula VII:

and pharmaceutically acceptable salts thereof, wherein 'p' is an integer from 5 to 20; and 'W' is phenyl or tert-butyl oxy. For instance, 'p' can be 10; and 'W' can be phenyl.

Multi-Linked Drug Conjugates



[0113] In yet another embodiment, a multi-drug conjugate is provided comprising a pharmaceutically active agent covalently bound to a plurality of stimuli-sensitive linkers, wherein each linker is covalently bound to at least one additional pharmaceutically active agent, wherein the conjugate releases at least one pharmaceutically active agent upon delivery to a target cell. Such conjugates can have a conformation similar to a dendrimer, and can comprise a series of conjugates in a chain.

[0114] In one aspect, the stimuli-sensitive linker can be a C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, or combinations thereof. For instance, the linker can be a C3 straight chain alkyl. In yet another instance, the linker can comprise a ketone.

[0115] In yet another aspect, the pharmaceutically active agent comprises anticancer chemotherapy agents. For instance, the pharmaceutically active agent can include doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.

Pharmaceutical Preparations and Methods of Administration



[0116] In another embodiment, a pharmaceutical composition is provided comprising the multi-drug conjugate above, or a pharmaceutically acceptable salt thereof, in a pharmaceutically acceptable vehicle.

[0117] The identified nanoparticles and multi-drug conjugates (i.e. compounds) treat, inhibit, control and/or prevent, or at least partially arrest or partially prevent, diseases that are treatable by known pharmaceutically active agents and can be administered to a subject at therapeutically effective doses for the inhibition, prevention, prophylaxis or therapy for such diseases. The compounds of the present invention comprise a therapeutically effective dosage of a nanoparticle and/or multi-drug conjugate, a term which includes therapeutically, inhibitory, preventive and prophylactically effective doses of the compounds of the present invention and is more particularly defined below. The subjects treated by administration of the compounds is preferably an animal, including, but not limited to, mammals, reptiles and avians, more preferably horses, cows, dogs, cats, sheep, pigs, and chickens, and most preferably human.

Therapeutically Effective Dosage



[0118] Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals for determining the LD50 (the dose lethal to 50% of the population) and the ED50, (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index that can be expressed as the ratio LD50/ED50. Compounds that exhibit large therapeutic indices are preferred. While compounds exhibiting toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site affected by the disease or disorder in order to minimize potential damage to unaffected cells and reduce side effects.

[0119] The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosages for use in humans and other mammals. The dosage of such compounds lies preferably within a range of circulating plasma or other bodily fluid concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dosage may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful dosages in humans and other mammals. Compound levels in plasma may be measured, for example, by high performance liquid chromatography.

[0120] The amount of a compound that may be combined with a pharmaceutically acceptable carrier to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. It will be appreciated by those skilled in the art that the unit content of a compound contained in an individual dose of each dosage form need not in itself constitute a therapeutically effective amount, as the necessary therapeutically effective amount could be reached by administration of a number of individual doses. The selection of dosage depends upon the dosage form utilized, the condition being treated, and the particular purpose to be achieved according to the determination of those skilled in the art.

[0121] The dosage regime for treating a disease or condition with the compounds of the invention is selected in accordance with a variety of factors, including the type, age, weight, sex, diet and medical condition of the patient, the route of administration, pharmacological considerations such as activity, efficacy, pharmacokinetic and toxicology profiles of the particular compound employed, and whether a compound delivery system is utilized. Thus, the dosage regime actually employed may vary widely from subject to subject.

Formulations and Use



[0122] The compounds of the present invention may be formulated by known methods for administration to a subject using several routes which include, but are not limited to, parenteral, oral, topical, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and ophthalmic routes. The individual compounds may also be administered in combination with one or more additional compounds of the present invention and/or together with other pharmaceutically active or inert agents. Such pharmaceutically active or inert agents may be in fluid or mechanical communication with the compound(s) or attached to the compound(s) by ionic, covalent, Van der Waals, hydrophobic, hydrophillic or other physical forces. It is preferred that administration is localized in a subject, but administration may also be systemic.

[0123] The compounds of the present invention may be formulated by any conventional manner using one or more pharmaceutically acceptable carriers. Thus, the compounds and their pharmaceutically acceptable salts and solvates may be specifically formulated for administration, e.g., by inhalation or insufflation (either through the mouth or the nose) or oral, buccal, parenteral or rectal administration. The compounds may take the form of charged, neutral and/or other pharmaceutically acceptable salt forms. Examples of pharmaceutically acceptable carriers include, but are not limited to, those described in REMINGTON'S PHARMACEUTICAL SCIENCES (A.R. Gennaro, Ed.), 21st edition, ISBN: 0781746736 (2005), incorporated herein by reference in its entirety.

[0124] The compounds may also take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, and the like. Such formulations will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.

Parenteral Administration



[0125] The compound may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form in ampoules or in multi-dose containers with an optional preservative added. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass, plastic or the like. The formulation may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.

[0126] For example, a parenteral preparation may be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent (e.g., as a solution in 1,3-butanediol). Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may be used in the parenteral preparation.

[0127] Alternatively, the compound may be formulated in powder form for constitution with a suitable vehicle, such as sterile pyrogen-free water, before use. For example, a compound suitable for parenteral administration may comprise a sterile isotonic saline solution containing between 0.1 percent and 90 percent weight per volume of the compound. By way of example, a solution may contain from about 5 percent to about 20 percent, more preferably from about 5 percent to about 17 percent, more preferably from about 8 to about 14 percent, and still more preferably about 10 percent of the compound. The solution or powder preparation may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection. Other methods of parenteral delivery of compounds will be known to the skilled artisan and are within the scope of the invention.

Oral Administration



[0128] For oral administration, the compound may take the form of tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents, fillers, lubricants and disintegrants:

A. Binding Agents



[0129] Binding agents include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof. Suitable forms of microcrystalline cellulose include, for example, the materials sold as AVICEL-PH-101, AVICEL-PH-103 and AVICEL-PH-105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, Pennsylvania, USA). An exemplary suitable binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581 by FMC Corporation.

B. Fillers



[0130] Fillers include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), lactose, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.

C. Lubricants



[0131] Lubricants include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laurate, agar, and mixtures thereof. Additional lubricants include, for example, a syloid silica gel (AEROSIL 200, manufactured by W.R. Grace Co. of Baltimore, Maryland, USA), a coagulated aerosol of synthetic silica (marketed by Deaussa Co. of Plano, Texas, USA), CAB-O-SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Massachusetts, USA), and mixtures thereof.

D. Disintegrants



[0132] Disintegrants include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.

[0133] The tablets or capsules may optionally be coated by methods well known in the art. If binders and/or fillers are used with the compounds of the invention, they are typically formulated as about 50 to about 99 weight percent of the compound. In one aspect, about 0.5 to about 15 weight percent of disintegrant, and particularly about 1 to about 5 weight percent of disintegrant, may be used in combination with the compound. A lubricant may optionally be added, typically in an amount of less than about 1 weight percent of the compound. Techniques and pharmaceutically acceptable additives for making solid oral dosage forms are described in Marshall, SOLID ORAL DOSAGE FORMS, Modern Pharmaceutics (Banker and Rhodes, Eds.), 7:359-427 (1979). Other less typical formulations are known in the art.

[0134] Liquid preparations for oral administration may take the form of solutions, syrups or suspensions. Alternatively, the liquid preparations may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol or fractionated vegetable oils); and/or preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, flavoring, coloring, perfuming and sweetening agents as appropriate. Preparations for oral administration may also be formulated to achieve controlled release of the compound. Oral formulations preferably contain 10% to 95% compound. In addition, the compounds of the present invention may be formulated for buccal administration in the form of tablets or lozenges formulated in a conventional manner. Other methods of oral delivery of compounds will be known to the skilled artisan and are within the scope of the invention.

Controlled-Release Administration



[0135] Controlled-release (or sustained-release) preparations may be formulated to extend the activity of the compound and reduce dosage frequency. Controlled-release preparations can also be used to effect the time of onset of action or other characteristics, such as blood levels of the compound, and consequently affect the occurrence of side effects.

[0136] Controlled-release preparations may be designed to initially release an amount of a compound that produces the desired therapeutic effect, and gradually and continually release other amounts of the compound to maintain the level of therapeutic effect over an extended period of time. In order to maintain a near-constant level of a compound in the body, the compound can be released from the dosage form at a rate that will replace the amount of compound being metabolized and/or excreted from the body. The controlled-release of a compound may be stimulated by various inducers, e.g., change in pH, change in temperature, enzymes, water, or other physiological conditions or molecules.

[0137] Controlled-release systems may include, for example, an infusion pump which may be used to administer the compound in a manner similar to that used for delivering insulin or chemotherapy to specific organs or tumors. Typically, using such a system, the compound is administered in combination with a biodegradable, biocompatible polymeric implant that releases the compound over a controlled period of time at a selected site. Examples of polymeric materials include polyanhydrides, polyorthoesters, polyglycolic acid, polylactic acid, polyethylene vinyl acetate, and copolymers and combinations thereof. In addition, a controlled release system can be placed in proximity of a therapeutic target, thus requiring only a fraction of a systemic dosage.

[0138] As an example, an implantable metronomic infusion pump can be used for local delivery of the nanoparticles and multi-drug conjugates of the present invention. See, e.g., U.S. Patent Nos. 7,799,016, 7,799,012, 7,588,564, 7,575,574, and 7,569,051, each of which is incorporated herein by reference in its entirety. In this example, a magnetically controlled pump can be implanted into the brain of a patient and deliver the nanoparticles and multi-drug conjugates at a controlled rate corresponding to the specific needs of the patient. A flexible double walled pouch that is formed from two layers of polymer can be alternately expanded and contracting by magnetic solenoid. When contracted, the nanoparticles and multi-drug conjugates can be pushed out of the pouch through a plurality of needles. When the pouch is expanded, surrounding cerebral fluid is drawn into the space between the double walls of the pouch from which it is drawn through a catheter to an analyzer. Cerebral fluid drawn from the patient can be analyzed. The operation of the apparatus and hence the treatment can be remotely controlled based on these measurements and displayed through an external controller.

[0139] The compounds of the invention may be administered by other controlled-release means or delivery devices that are well known to those of ordinary skill in the art. These include, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, or a combination of any of the above to provide the desired release profile in varying proportions. Other methods of controlled-release delivery of compounds will be known to the skilled artisan and are within the scope of the invention.

Inhalation Administration



[0140] The compound may also be administered directly to the lung by inhalation. For administration by inhalation, a compound may be conveniently delivered to the lung by a number of different devices. For example, a Metered Dose Inhaler ("MDI") which utilizes canisters that contain a suitable low boiling point propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas may be used to deliver a compound directly to the lung. MDI devices are available from a number of suppliers such as 3M Corporation, Aventis, Boehringer Ingleheim, Forest Laboratories, Glaxo-Wellcome, Schering Plough and Vectura.

[0141] Alternatively, a Dry Powder Inhaler (DPI) device may be used to administer a compound to the lung. DPI devices typically use a mechanism such as a burst of gas to create a cloud of dry powder inside a container, which may then be inhaled by the patient. DPI devices are also well known in the art and may be purchased from a number of vendors which include, for example, Fisons, Glaxo-Wellcome, Inhale Therapeutic Systems, ML Laboratories, Qdose and Vectura. A popular variation is the multiple dose DPI ("MDDPI") system, which allows for the delivery of more than one therapeutic dose. MDDPI devices are available from companies such as AstraZeneca, GlaxoWellcome, IVAX, Schering Plough, SkyePharma and Vectura. For example, capsules and cartridges of gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch for these systems.

[0142] Another type of device that may be used to deliver a compound to the lung is a liquid spray device supplied, for example, by Aradigm Corporation. Liquid spray systems use extremely small nozzle holes to aerosolize liquid compound formulations that may then be directly inhaled into the lung. For example, a nebulizer device may be used to deliver a compound to the lung. Nebulizers create aerosols from liquid compound formulations by using, for example, ultrasonic energy to form fine particles that may be readily inhaled. Examples of nebulizers include devices supplied by Sheffield/Systemic Pulmonary Delivery Ltd., Aventis and Batelle Pulmonary Therapeutics.

[0143] In another example, an electrohydrodynamic ("EHD") aerosol device may be used to deliver a compound to the lung. EHD aerosol devices use electrical energy to aerosolize liquid compound solutions or suspensions. The electrochemical properties of the compound formulation are important parameters to optimize when delivering this compound to the lung with an EHD aerosol device. Such optimization is routinely performed by one of skill in the art. Other methods of intra-pulmonary delivery of compounds will be known to the skilled artisan and are within the scope of the invention.

[0144] Liquid compound formulations suitable for use with nebulizers and liquid spray devices and EHD aerosol devices will typically include the compound with a pharmaceutically acceptable carrier. In one exemplary embodiment, the pharmaceutically acceptable carrier is a liquid such as alcohol, water, polyethylene glycol or a perfluorocarbon. Optionally, another material may be added to alter the aerosol properties of the solution or suspension of the compound. For example, this material may be a liquid such as an alcohol, glycol, polyglycol or a fatty acid. Other methods of formulating liquid compound solutions or suspensions suitable for use in aerosol devices are known to those of skill in the art.

Depot Administration



[0145] The compound may also be formulated as a depot preparation. Such long-acting formulations may be administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection. Accordingly, the compounds may be formulated with suitable polymeric or hydrophobic materials such as an emulsion in an acceptable oil or ion exchange resins, or as sparingly soluble derivatives such as a sparingly soluble salt. Other methods of depot delivery of compounds will be known to the skilled artisan and are within the scope of the invention.

Topical Administration



[0146] For topical application, the compound may be combined with a carrier so that an effective dosage is delivered, based on the desired activity ranging from an effective dosage, for example, of 1.0 µM 1.0 mM. In one aspect of the invention, a topical compound can be applied to the skin. The carrier may be in the form of, for example, and not by way of limitation, an ointment, cream, gel, paste, foam, aerosol, suppository, pad or gelled stick.

[0147] A topical formulation may also consist of a therapeutically effective amount of the compound in an ophthalmologically acceptable excipient such as buffered saline, mineral oil, vegetable oils such as corn or arachis oil, petroleum jelly, Miglyol 182, alcohol solutions, or liposomes or liposome-like products. Any of these compounds may also include preservatives, antioxidants, antibiotics, immunosuppressants, and other biologically or pharmaceutically effective agents which do not exert a detrimental effect on the compound. Other methods of topical delivery of compounds will be known to the skilled artisan and are within the scope of the invention.

Suppository Administration



[0148] The compound may also be formulated in rectal formulations such as suppositories or retention enemas containing conventional suppository bases such as cocoa butter or other glycerides and binders and carriers such as triglycerides, microcrystalline cellulose, gum tragacanth or gelatin. Suppositories can contain the compound in the range of 0.5% to 10% by weight. Other methods of suppository delivery of compounds will be known to the skilled artisan and are within the scope of the invention.

Other Systems of Administration



[0149] Various other delivery systems are known in the art and can be used to administer the compounds of the invention. Moreover, these and other delivery systems may be combined and/or modified to optimize the administration of the compounds of the present invention.

Ratiometric Control of Drug-Linker and Drug-Drug Compositions in a Nanoparticle



[0150] In yet another embodiment, a method is provided for controlling ratios of conjugated drugs contained in a nanoparticle inner sphere, the method comprising: a) synthesizing a combination of a first drug independently conjugated to a stimuli-sensitive linker, and a second drug independently conjugated to a linker having the same composition, wherein the first drug conjugate and second drug conjugate have a predetermined ratio; b) adding the combination to an agitated solution comprising a polar lipid; and c) adding water to the agitated solution, wherein nanoparticles are produced having a controlled ratio of conjugated drugs contained in the inner sphere. Unlike other methods that require several additional steps to create nanoparticles, the present self assembly of the nanoparticles containing combinations of conjugated drugs is highly efficient.

[0151] In one aspect, the first drug and the second drug can independently include an antibiotic, antimicrobial, antiviral, growth factor, chemotherapeutic agent, and combinations thereof. For instance, the first drug and the second drug are independently selected from the group consisting of doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.

[0152] In another aspect, the stimuli-sensitive linker is a pH-sensitive linker. For instance, the stimuli-sensitive linker is selected from the group consisting of C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, and combinations thereof.

[0153] In various aspects of the present embodiment, the combination of conjugated drugs having a predetermined ratio further comprises at least one additional drug independently conjugated to a stimuli-sensitive linker having the same composition.

[0154] In yet another embodiment, a method is provided for controlling ratios of conjugated drugs contained in a nanoparticle inner sphere, the method comprising: a) synthesizing a combination of (i) a first drug and a second drug conjugated by a first stimuli-sensitive linker, and (ii) a first drug and a second drug conjugated by a second stimuli-sensitive linker, wherein the first drug conjugate and second drug conjugate have a predetermined ratio; b) adding the combination to an agitated solution comprising a polar lipid; and c) adding water to the agitated solution, wherein nanoparticles are produced having a controlled ratio of conjugated drugs contained in the inner sphere.

[0155] In one aspect, the first drug and the second drug are independently selected from the group consisting of an antibiotic, antimicrobial, antiviral, growth factor, chemotherapeutic agent, and combinations thereof. For instance, the first drug and the second drug can independently include doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.

[0156] In another aspect, the stimuli-sensitive linker is a pH-sensitive linker. For instance, the first stimuli-sensitive linker and the second stimuli-sensitive linker can independently include C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, and combinations thereof.

[0157] In various aspects of the present embodiment, the combination of conjugated drugs having a predetermined ratio further comprises at least one additional conjugate of a first drug and a second drug conjugated by a stimuli-sensitive linker other than those present in the combination.

Methods of Synthesizing Drug-Linker and Drug-Drug Conjugate Containing Nanoparticles



[0158] In another embodiment, a method is provided for producing a combination of conjugated drugs having a predetermined ratio in a nanoparticle, said nanoparticle comprising an inner sphere, the method comprising: a) adding to an agitated solution comprising a polar lipid a combination of a first drug independently conjugated to a stimuli-sensitive linker, and a second drug independently conjugated to a linker having the same composition, wherein the first drug conjugate and the second drug conjugate have a predetermined ratio; and b) adding water to the agitated solution, wherein nanoparticles are produced containing in the inner sphere the conjugated drugs having a predetermined ratio. In various aspects, the method can further comprise: c) isolating nanoparticles having a diameter less than about 300 nm.

[0159] In various aspects, the first drug and the second drug are independently selected from the group consisting of an antibiotic, antimicrobial, growth factor, chemotherapeutic agent, and combinations thereof. For instance, the first drug and the second drug can independently include doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.

[0160] In yet another aspect, the stimuli-sensitive linker is a pH-sensitive linker. For instance, the stimuli-sensitive linker can be C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, or combinations thereof.

[0161] In yet another aspect, the combination of conjugated drugs having a predetermined ratio further comprise a third drug independently conjugated to a stimuli-sensitive linker having the same composition. In various aspects, the solution comprising a polar lipid further comprises a functionlalized polar lipid.

[0162] In yet another embodiment, a method is provided for producing a combination of conjugated drugs having a predetermined ratio in a nanoparticle, said nanoparticle comprising an inner sphere, the method comprising: a) adding to an agitated solution comprising a polar lipid a combination of (i) a first drug and second drug conjugated by a first stimuli-sensitive linker, and (ii) a first drug and a second drug conjugated by a second stimuli-sensitive linker, wherein the first drug conjugate and second drug conjugate have a predetermined ratio; and b) adding water to the agitated solution, wherein nanoparticles are produced containing in the inner sphere the conjugated drugs having a predetermined ratio. In various aspects, the method can further comprise: c) isolating nanoparticles having a diameter less than about 300 nm.

[0163] In one aspect, the first drug and the second drug can independently include an antibiotic, antimicrobial, growth factor, chemotherapeutic agent, and combinations thereof. For instance, the first drug and the second drug are independently selected from the group consisting of doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.

[0164] In another aspect, the stimuli-sensitive linker is a pH-sensitive linker. For instance, the first stimuli-sensitive linker and the second stimuli-sensitive linker can independently be C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, and combinations thereof.

[0165] In various aspects of the present embodiment, the combination of conjugated drugs having a predetermined ratio further comprises at least one additional conjugate of a first drug and a second drug conjugated by a stimuli-sensitive linker other than those present in the combination. In various aspects, the solution comprising a polar lipid further comprises a functionlalized polar lipid. An example of a polar lipid is a phospholipid as defined herein.

Methods of Treating Diseases and Conditions in a Subject



[0166] The pharmaceutically active agents used in the present invention are known to provide a certain response when administered to subjects. One of skill in the art will readily be able to choose particular pharmaceutically active agents to use with the nanoparticles and multi-drug conjugates to treat certain diseases or conditions, including those listed in the appended tables. In addition, the literature is replete with examples of administering pharmaceutically active agents to subjects, especially those regulated by the government.

[0167] Therefore, a method is provided for treating a disease or condition, the method comprising administering a therapeutically effective amount of the nanoparticle above to a subject in need thereof. In one aspect, the disease is a proliferative disease including lymphoma, renal cell carcinoma, prostate cancer, lung cancer, pancreatic cancer, melanoma, colorectal cancer, ovarian cancer, breast cancer, glioblastoma multiforme and leptomeningeal carcinomatosis. In another aspect, the disease is a heart disease including Atherosclerosis, Ischemic heart disease, Rheumatic heart disease, Hypertensive heart disease, Infective endocarditis, Coronary heart disease, and Constrictive pericarditis. In another aspect, the disease is an ocular disease selected from the group consisting of macular edema, retinal ischemia, macular degeneration, uveitis, blepharitis, keratitis, rubeosis iritis, iridocyclitis, conjunctivitis, and vasculitis. In another aspect, the disease is a lung disease including asthma, Chronic Bronchitis, Cystic Fibrosis, Emphysema, Pneumonia, lung cancer, Primary Pulmonary Hypertension, Pulmonary Arterial Hypertension, and Tuberculosis. In yet another aspect, the disease includes bacterial infection, viral infection, fungal infection, and parasitic infection.

[0168] In various aspects of the present embodiment, the nanoparticle is administered systemically. In another aspect, the nanoparticle is administered locally. In yet another aspect, the local administration is via implantable metronomic infusion pump.

[0169] In yet another embodiment, a method is provided for treating a disease or condition, the method comprising administering a therapeutically effective amount of the multi-drug conjugate above to a subject in need thereof. In one aspect, the disease is a proliferative disease including lymphoma, renal cell carcinoma, prostate cancer, lung cancer, pancreatic cancer, melanoma, colorectal cancer, ovarian cancer, breast cancer, glioblastoma multiforme and leptomeningeal carcinomatosis. In one aspect, the disease is a heart disease including Atherosclerosis, Ischemic heart disease, Rheumatic heart disease, Hypertensive heart disease, Infective endocarditis, Coronary heart disease, and Constrictive pericarditis. In one aspect, the disease is an ocular disease including macular edema, retinal ischemia, macular degeneration, uveitis, blepharitis, keratitis, rubeosis iritis, iridocyclitis, conjunctivitis, and vasculitis. In one aspect, the disease is a lung disease including asthma, Chronic Bronchitis, Cystic Fibrosis, Emphysema, Pneumonia, lung cancer, Primary Pulmonary Hypertension, Pulmonary Arterial Hypertension, and Tuberculosis. In yet another aspect, the disease is selected from the group consisting of bacterial infection, viral infection, fungal infection, and parasitic infection.

[0170] In various aspects of the present embodiment, the multi-drug conjugate is administered systemically. In another aspect, the multi-drug conjugate is administered locally. In yet another aspect, the local administration is via implantable metronomic infusion pump.

Methods of Sequentially Delivering a Pharmaceutically Active Drug to a Target



[0171] In yet another embodiment, a method is provided for sequentially delivering a drug conjugate to a target cell. Preferably, a combination of drug-drug conjugates having individual linkers of varying sensitivities is administered in an environment whereby one individual linker is triggered first, followed by another individual linker triggered at another condition. Therefore, the method comprises administering a nanoparticle above to the target cell and triggering multi-drug conjugate release. In various aspects of the present embodiment, the nanoparticle is administered systemically. In another aspect, the nanoparticle is administered locally. In yet another aspect, the local administration is via implantable metronomic infusion pump.

Methods of Nanoencapsulation with High Loading Efficiency



[0172] In yet another embodiment, a method is provided for nanoencapsulation of a plurality of drugs comprising: separately linking each of the plurality of drugs with a corresponding polymer backbone with nearly 100% loading efficiency by forming the corresponding polymer backbone by ring opening polymerization beginning with the corresponding drug, wherein each of the corresponding polymer backbones has the same or similar physicochemical properties and has approximately the same chain length; mixing the plurality of linked drugs and polymers at selectively predetermined ratios at selectively and precisely controlled drug ratios; and synthesizing the mixed plurality of linked drugs and polymers into a nanoparticle.

[0173] In various aspects, the plurality of drugs can independently include an antibiotic, antimicrobial, growth factor, chemotherapeutic agent, and combinations thereof. For instance, the plurality of drugs can independently include doxorubicin, camptothecin, gemicitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansanol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.

[0174] In various aspects, the polymer backbone is a stimuli-sensitive linker. For instance, the stimuli-sensitive linker can include a C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, and combinations thereof.

Kits



[0175] In various embodiments, the present invention can also involve kits. Such kits can include the compounds of the present invention and, in certain embodiments, instructions for administration. When supplied as a kit, different components of a compound formulation can be packaged in separate containers and admixed immediately before use. Such packaging of the components separately can, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the compound. The pack may, for example, comprise metal or plastic foil such as a blister pack. Such packaging of the components separately can also, in certain instances, permit long-term storage without losing activity of the components. In addition, if more than one route of administration is intended or more than one schedule for administration is intended, the different components can be packaged separately and not mixed prior to use. In various embodiments, the different components can be packaged in one combination for administration together.

[0176] Kits may also include reagents in separate containers such as, for example, sterile water or saline to be added to a lyophilized active component packaged separately. For example, sealed glass ampules may contain lyophilized compounds and in a separate ampule, sterile water, sterile saline or sterile each of which has been packaged under a neutral non-reacting gas, such as nitrogen. Ampules may consist of any suitable material, such as glass, organic polymers, such as polycarbonate, polystyrene, ceramic, metal or any other material typically employed to hold reagents. Other examples of suitable containers include bottles that may be fabricated from similar substances as ampules, and envelopes that may consist of foil-lined interiors, such as aluminum or an alloy. Other containers include test tubes, vials, flasks, bottles, syringes, and the like. Containers may have a sterile access port, such as a bottle having a stopper that can be pierced by a hypodermic injection needle. Other containers may have two compartments that are separated by a readily removable membrane that upon removal permits the components to mix. Removable membranes may be glass, plastic, rubber, and the like.

[0177] In certain embodiments, kits can be supplied with instructional materials. Instructions may be printed on paper or other substrate, and/or may be supplied as an electronic-readable medium, such as a floppy disc, mini-CD-ROM, CD-ROM, DVD-ROM, Zip disc, videotape, audio tape, and the like. Detailed instructions may not be physically associated with the kit; instead, a user may be directed to an Internet web site specified by the manufacturer or distributor of the kit, or supplied as electronic mail.

EXAMPLES



[0178] Aspects of the present teachings may be further understood in light of the following examples, which should not be construed as limiting the scope of the present teachings in any way.

Example 1- Ratiometric Combinatorial Drug and Nanoparticle Synthesis


Materials.



[0179] L-lactide was purchased from Sigma-Aldrich Co. (Milwaukee, WI), recrystallized three times in ethylacetate and dried under vacuum. L-lactide crystals were further dried inside a glove box and sealed into a glass vial under dry argon and then stored at -20 °C prior to use. 2,6-di-iso-propylaniline (Sigma-Aldrich Co.) and 2,4-pentanedione (Alfa Aesar Co., Ward Hill, MA) were used as received. All other chemicals and anhydrous solvents were purchased from Sigma-Aldrich Co. unless otherwise specified. Anhydrous tetrahydrofuran (THF) and toluene were prepared by distillation under sodium benzophenone and were kept anhydrous by using molecular sieves. The 2-((2,6-diisopropylphenyl)amino)-4-((2,6-diisopropylphenyl)imino)-2-pentene (BDI) ligand and the corresponding metal catalysts (BDI)ZnN(SiMe3)2 were prepared inside a glove box following a published protocol and stored at -20 °C prior to use (B. M. Chamberlain, M. Cheng, D. R. Moore, T. M. Ovitt, E. B. Lobkovsky, G. W. Coates, J Am Chem Soc 2001, 123, 3229-3238). DOX·HCl was purchased from Jinan Wedo Co., Ltd. (Jinan, China) and used as received. Removal of HCl from DOX·HCl was achieved by neutralizing DOX·HCl solution in water with triethyleamine, after which the solution color changed from red to purple. The free base form of DOX was subsequently extracted with dichloromethane. The organic extract was filtered through anhydrous Na2SO4 and dried under vacuum to collect DOX crystals. (S)-(+)-Camptothecine (CPT) was purchased from TCI America and used as received.

Synthesis of 2-((2,6-diisopropylphenyl)amino)-4-((2,6-diisopropylphenyl)imino)-2-pentene (BDI).



[0180] Ligand BDI was prepared following a previously published protocol with minor modification (B. M. Chamberlain, M. Cheng, D. R. Moore, T. M. Ovitt, E. B. Lobkovsky, G. W. Coates, J Am Chem Soc 2001, 123, 3229-3238). Briefly, 2,6-Di-n-propylaniline (13.0 mmol) and 2,4-pentanedione (6.5 mmol) in the ratio of 2:1 were dissolved in absolute ethanol (20 ml). The mixture solution was acidified with concentrated HCl (0.6 mL) and heated at reflux for 48 h, which resulted in white precipitates. After being cooled to room temperature, the white precipitates were dissolved with dichloromethane and saturated aqueous bicarbonate solution. The orange colored solution was then extracted and washed with brine three times and filtered through anhydrous Na2SO4, followed by being concentrated and precipitated in hexane. The resulting precipitates were collected by filtration, suspended in diethyl ether (20 mL), and washed with saturated aqueous bicarbonate followed by brine. The organic layer was then separated through filtration in the presence of Na2SO4 to absorb moisture and then precipitated in hexane as a light brown powder (yield ~ 60%). 1H NMR (JEOL, CDCl3, 500 MHz): δ 12.20 (br, 1H, NH), 7.12 (m, 6H, ArH), 4.83 (s, 1H, Hβ), 3.10 (m, 4H, CHMe2), 1.72 (s, 6H, α-Me), 1.22 (d, 12H, CHMeMe), 1.12 (d, 12H, CHMeMe) ppm. ESI-MS (positive): m/z = 419.43 [M+H]+.

Synthesis of (BDI)ZnN(SiMe3)2 catalyst.



[0181] Zinc bis-(trimethylsilyl)amide (463 mg, 1.19 mmol) in toluene (20 mL) was added into a solution of BDI (500 mg, 1.19 mmol) in toluene (20 mL). The mixture solution was stirred for 18 h at 80 °C and then the solvent was removed under vacuum to form (BDI)ZnN(SiMe3)2 as a light yellow solid, which was recrystallized from toluene at -30 °C to yield colorless blocks (yield ~ 70%).1H NMR (JEOL, C6D6, 500 MHz): δ (br, 1H, NH), 6.9-7.13 (m, 6H, ArH), 4.85 (s, 1H,Hβ), 3.25 (m, 4H, CHMe2), 1.67 (s, 6H, α-Me), 1.1-1.25 (d, 12H+12H=24H, CHMeMe), 0.08-0.1 (18H, s, SiCH3) ppm.

Ring opening polymerization of l-lactide.



[0182] Following previously published protocols, DOX-PLA and CPT-PLA polymers were synthesized through ring opening polymerization of l-lactide initiated by alkoxy complex of (BDI)ZnN(SiMe3)2 in a glove box under argon environment at room temperature. For the synthesis of DOX-PLA, (BDI)ZnN(SiMe3)2 (6.4 mg, 0.01 mmol) and DOX (5.4 mg, 0.01 mmol) were mixed in 0.5 mL of anhydrous THF. L-lactide (101.0 mg, 0.7 mmol) dissolved in 2 mL anhydrous THF was added dropwise. After the l-lactide was completely consumed, the crude product was precipitated in cold diethyl ether, yielding DOX-PLA conjugates. The CPT-PLA conjugates were synthesized in the same procedures as the DOX-PLA. These drug-polymer conjugates had a molecular weight of about 10,000 g/mole determined by gel permeation chromatography.

Synthesis of lipid-coated drug-polymer conjugate nanoparticles.



[0183] Lipid-polymer hybrid nanoparticles with polymeric cores consisting of the synthesized drug-polymer conjugates were prepared through a nanoprecipitation method (L. Zhang, J. M. Chan, F. X. Gu, J. W. Rhee, A. Z. Wang, A. F. Radovic-Moreno, F. Alexis, R. Langer, O. C. Farokhzad, ACS Nano 2008, 2, 1696-1702). In detail, 200 ug of egg PC (Avanti Polar Lipids Inc.) and 260 ug of 1,2- distearoyl-sn-glycero-3- phosphoethanolamine-N-carboxy(polyethyleneglycol)-2000 (DSPE-PEG-COOH) (Avanti Polar Lipids Inc.) were dissolved in 4% ethanol and stirred and heated at 68 °C for 3 min. A total of 500 ug of DOX-PLA and CPT-PLA was dissolved in acetonitrile and added dropwise to the lipid solution while stirring. The solution was then vortexed for 3 min followed by the addition of deionized water (1 mL). Then the diluted solution was stirred at room temperature for 2 h, washed with PBS buffer using an Amicon Ultra centrifugal filter with a molecular weight cutoff of 100 kDa (Millipore, Billerica, MA), and resuspended in 1mL of PBS. Nanoparticles with different DOX/CPT drug ratios were prepared by adjusting the amount of each type of drug-polymer conjugates while keeping the total polymer weight at 500 ug. The nanoparticle size and surface zeta potential were obtained from three repeat measurements by dynamic light scattering (DLS) (Malvern Zetasizer, ZEN 3600) with a backscattering angle of 173°. The morphology of the particles was characterized by scanning electron microscopy (SEM) (Phillips XL30 ESEM). Samples for SEM were prepared by dropping nanoparticle solution (5 µL) onto a polished silicon wafer. After drying the droplet at room temperature overnight, the sample was coated with chromium and then imaged by SEM. The drug loading yield of the synthesized nanoparticles was determined by UV-spectroscopy (TECAN, infinite M200) using the maximum absorbance at 482 nm for DOX and 362 nm for CPT. No shift in the absorbance peak was observed between the free drugs and their polymer conjugates. Standard calibration curves of both DOX and CPT at various concentrations were obtained to quantify drug concentrations in the nanoparticles.

Cellular colocalization and cytotoxicity studies.



[0184] The MDA-MB-435 cell line was maintained in Dulbecco's modification of Eagle's medium (DMEM, Mediatech, Inc.) supplemented with 10% fetal calf albumin, penicillin/streptomycin (GIBCO®), L-glutamine (GIBCO®), nonessential amino acids, sodium bicarbonate, and sodium pyruvate (GIBCO®). The cells were cultured at 37°C and 5% CO2. For the dual-drug colocalization and cellular internalization study, the cells were incubated with dual-drug loaded nanoparticles for 4 h, washed with PBS, and fixed on a chamber slide for fluorescence microscopy imaging. The cytotoxicity of the dual-drug loaded nanoparticles was assessed using the (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay (Promega, Madison, WI). Briefly, the cells were seeded at 25% confluency (∼4×103 cells/well) in 96-well plates and incubated with different concentrations of drug loaded nanoparticles for 24 h. The cells were then washed with PBS three times and incubated in fresh media for an additional 72h. MTT assay was then applied to the samples to measure the viability of the cells following the manufacturer's instruction.

Results



[0185] In the study, we used (BDI)ZnN(SiMe3)2, a metal-amido complex in which BDI refers to 2-((2,6-diisopropylphenyl)amido)-4-((2,6diisopropylphenyl) -imino)-2-pentene, as a catalyst for the in-situ formation of metal-alkoxide with the hydroxyl group of DOX and CPT to initiate the living polymerization of l-lactide and form drug-poly-l-lactide (drug-PLA) conjugates (Figure 2A). The formation of the drug-polymer conjugates was verified by the 1H-NMR spectroscopy, which exhibits all the characteristic proton resonance peaks corresponding to the parent drug molecules. The appearance of the aromatic proton resonance at δ 7.5 to 8.0 ppm in DOX-PLA conjugates (Figure 2B, top panel) and δ 7.5 to 8.5 ppm in CPT-PLA conjugates (Figure 2B, bottom panel) along with the characteristic -CH3 proton of PLA at δ 1.5 ppm and -CH proton at δ 5.2 ppm confirms the formation of the drug-polymer conjugates. The desired drug-polymer conjugation products were further validated by gel permeation chromatography (GPC) which shows the molecular weight as 10,000 Dalton for both DOX-PLA and CPT-PLA conjugates (Figure 2C). The molecular weight is in accord with the monomer-to-initiator feed ratio which indicates near 100% conversion of the monomers to polymers. Since the formation of metal alkoxide complex is quantitative and the reaction is homogeneous, the reaction proceeded quantitatively such that all monomers were converted into products. Also the molecular weight of the polymer matches that from an earlier study conducted by Tong et al. who used (BDI)ZnN(SiMe3)2 to catalyze the ring opening polymerization of both DOX and CPT. (R. Tong, J. Cheng, Bioconjug Chem 2010, 21, 111-121; R. Tong, J. Cheng, J Am Chem Soc 2009, 131, 4744-4754).

[0186] Upon successful synthesis of the drug-polymer conjugates, we used them to prepare lipid-polymer hybrid nanoparticles for dual-drug delivery. Using DSPE-PEG and phospholipids to coat the polymeric nanoparticle core, the resulting lipid-polymer hybrid nanoparticles are highly stable in water, PBS and serum and have high drug loading yield as the entire polymeric core consists of the drug-polymer conjugates. Moreover, by simply adjusting the DOX-PLA:CPT-PLA molar ratio, dual-drug loaded nanoparticles with ratiometric drug loading of DOX and CPT were prepared. Keeping the total drug-polymer conjugates weight constant at 1mg, we varied the DOX-PLA:CPT-PLA ratio to tune the ratiometric drug loading. The resulting drug-loaded nanoparticles exhibit a unimodal size distribution at ∼100 nm with low PDI values (Figure 3). In addition, the particles possess negative surface zeta potential, which is consistent with the DSPE-PEG-COOH coating and serves to prevent the particles from aggregation. The particle size measured by DLS was consistent with the SEM images of the particles (Figure 3).

[0187] Following the physicochemical characterization of the particles, we next examined the drug loading efficiency in these drug-polymer conjugate nanoparticle systems. We prepared various formulations of the nanoparticles with different ratios of drug-polymer conjugates and found that, in all cases, over 90% of the conjugates were encapsulated into the nanoparticles (Figure 4). No change in loading efficiency was observed when DOX-PLA and CPT-PLA conjugates were loaded in combination or separately, presumably due to the fact that the long and sharply distributed PLA polymer chain gives each drug molecule a predominant and uniform hydrophobic property. Therefore, they were completely encapsulated and stabilized by the lipid and the lipid-PEG layers in the lipid-polymer hybrid nanoparticle system. Furthermore, we varied the DOX-PLA: CPT-PLA molar ratios from 1:1, to 3:1 and to 1:3, while keeping the total drug-polymer conjugates mass constant. It was found that the final loading yields of DOX and CPT in the dual-drug loaded nanoparticles were highly consistent with the initial DOX-PLA: CPT-PLA molar ratios (supporting information the following table).
TABLE 1 - Characteristic features of the lipid-coated drug-polymer conjugate nanoparticles
DOX-PLA/CPT-PLA molar ratios 1:0 0:1 1:1 3:1 1:3
Particle size (nm)     100±2    
Particle PDI     0.17-0.22    
Particle zeta potential (mV)     -47±2    
DOX loading (µM) 47.8±0.2 0 24.0+0.1 35.8±0.2 12.0±0.8
CPT loading (µM) 0 48.2±0.1 24.4±0.1 12.3±0.1 36.2±0.2


[0188] These results further confirm that this approach enables one to encapsulate different types of drugs to the same nanoparticles with ratiometric control over drug loading.

[0189] Upon verifying the excellent drug loading efficiency in the present system, we then examined whether the different drug-polymer conjugates are loaded into the same nanoparticles as opposed to forming two different particle populations. To this end, we studied the colocalization of the two drug molecules and their internalization into cells through fluorescence microscopy. Since DOX is also a highly fluorescent molecule, the DOX-PLA conjugates can be identified from DOX's characteristic fluorescence wavelength (excitation/emission = 540 nm/600 nm). To visualize CPT-PLA, we attached a fluorescent probe, 6-((7-amino-4-mehylcoumarin-3-acetyl) amino) hexanoic acid succinimidyl ester (excitation/emission = 353 nm/442 nm), to the hydroxyl end of the CPT-PLA. Figure 5A shows the fluorescence microscopy images that exhibit the colocalization of the DOX-PLA and the CPT-PLA-probe. The colocalization study indicates that no segregation between the two types of drug-polymer conjugates occurs and each particle contains both DOX and CPT.

[0190] After having confirmed that the nanoparticles contain a mixture of DOX and CPT, we next examined the cytotoxicity of these dual-drug loaded nanoparticles in comparison to the cocktail mixtures of the corresponding single-drug loaded nanoparticles against MDA-MB-435 breast cancer cells in vitro. The cocktail system was prepared by mixing DOX-PLA loaded nanoparticles and CPT-PLA loaded nanoparticles at a ratio that is equivalent to the DOX-PLA:CPT-PLA molar ratio in the dual-drug nanoparticles. Figure 5B shows the results of IC50 measurements of the dual-drug loaded nanoparticles and cocktail combination of single-drug loaded nanoparticles. It was found that the dual-drug loaded nanoparticles consistently showed higher potency as compared to the cocktail systems for the 3 different drug ratios. In the 3:1, 1:1, and 1:3 DOX-PLA:CPT-PLA combinations, the dual-drug loaded nanoparticles showed an enhancement in efficacy by 3.5, 2.5, and 1.1 times, respectively, compared to the cocktail particle mixtures. This enhanced cytotoxicity of the dual-drug delivery system can be explained, at least partially, by the fact that dual-drug loaded nanoparticles can deliver more consistent combination drug payloads when compared to cocktail nanoparticle systems and hence maximize their combinatorial effect. In the cocktail mixture, variations in the nanoparticle uptake and the random drug distribution in cells likely compromised the efficacy of the drug combinations. Figure 5 suggests that the dual-drug loaded nanoparticles enable concurrent combination drug delivery through particle endocytosis. Once engulfed by the plasma membrane, nanoparticles are transported by endosomal vesicles before unloading their drug payloads. This endocytic uptake mechanism is particularly favourable to the drug-polymer conjugate system used in the present combinatorial drug delivery scheme. The pH drop associated with endosome maturation subjects the nanoparticles to an acidic environment and enzymatic digestions, which facilitate the cleavage of the ester linkage between the drug and the polymers. In addition, the degradation of the polymer PLA releases lactic acid to further lower the pH surrounding the nanoparticles, thereby further accelerating the drug release.

[0191] In conclusion, a new and robust approach for combination chemotherapy was presented by incorporating two different types of drugs with ratiometric control over drug loading into a single polymeric nanoparticle. By adapting metal alkoxide chemistry, drug conjugated polymers were synthesized in a quantitative yield with 100% monomer conversion, resulting in the formation of highly hydrophobic drug-polymer conjugates. These drug-polymer conjugates were successfully encapsulated into lipid-coated polymeric nanoparticles with over 90% loading efficiency. Using DOX and CPT as two model chemotherapy drugs, various ratios of DOX-PLA and CPT-PLA were loaded into the nanoparticles, yielding particles that are uniform in size, size distribution and surface charge. The cytotoxicity of these dual-drug carrying nanoparticles was compared with their cocktail their cocktail mixtures of single-drug loaded nanoparticles and showed superior therapeutic effect. This strategy can also be exploited for various other chemotherapeutic agents containing hydroxyl groups as well as different types of combinations for combinatorial treatments of various diseases. While only two drugs (DOX and CPT) were used to demonstrate the concept of this combinatorial drug delivery approach, this method can be generalized to incorporate three or more different types of drugs into the same nanoparticles with ratiometro control over drug loading.

Example 2 - Synthesis of Multi-Drug Conjugates


Synthesis of PTXL-GEM Conjugates



[0192] Paclitaxel (PTXL) and Gemcitabine hydrochloride (GEM) were purchased from ChemiTek Company and used without further purification. All other materials including solvents were purchased from Sigma-Aldrich Company, USA. Single addition luminescence ATP detection assay for cytotoxicity measurement was purchased from PerkinElmer Inc. 1H NMR spectra were recorded in CDCl3 using a Varian Mercury 400 MHz spectrometer. Electrospray ionization mass spectrometry (ESI-MS, Thermo LCQdeca mass spectrometer) and Thermo Fisher Scientific LTQ-XL Orbitrap mass spectrometer were used to determine the mass and molecular formula of the compounds, respectively. Reversed phase HPLC purification was performed on an Varian HPLC system equipped with µ-bonapack C18 column (4.6 mm × 150 mm, Waters Associates, Inc.) using acetonitrile and water (50/50, v/v) as mobile phase. Thin-layer chromatography (TLC) measurements were carried out using pre-coated silica gel HLF250 plates (Advenchen Laboratories, LLC, USA). 4-(N,N-dimethylamino) pyridinium-4-toluenesulfonate (DPTS) was prepared by mixing saturated THF solutions of N,N-dimethylaminopyridine (DMAP) (1 equiv) and p-toluenesulfonic acid monohydrate (1 equiv) at room temperature. The precipitate was filtered, washed three times with tetrahydrofuran (THF), and dried under vacuum.

Synthesis of compound 1.



[0193] Paclitaxel (5 mg, 5.8 µmol) and glutaric anhydride (2 mg. 17.5 µmol) were dissolved in 200 µL dry pyridine. To this solution, DMAP (0.57 µmol) dissolved in 10 µL pyridine was added and the solution was stirred at room temperature for 3 hrs. The reaction was monitored by TLC using 9.2/0.8 (v/v) CHCl3/MeOH as an eluent (product Rf = 0.42). The complete disappearance of the starting paclitaxel (Rf = 0.54) occurred after 3 hrs of reaction. Then the reaction was quenched by diluting the solution with dichloromethane (DCM), followed by extracting DMAP and pyridine with DI water. The remaining dichloromethane solution was concentrated and precipitated in hexane, resulting in 5.1 mg of the compound 1 as a white powder. The production yield was about 90%. 1H NMR (CDCl3, δ ppm) was carried out to characterize the produced compound 1 (Figure 15): 1.14 (s, 3H), 1.25 (s, 3H), 1.69 (s, 3H), 1.9-2.06 (broad, 7H), 2.16-2.27 (br, 4H), 2.2-2.7 (br, 14 H), 3.82 (d, 1H), 4.21 (d,1H), 4.32 (d-1H), 4.48 (t, 1H), 5.0 (d, 1H), 5.5 (d, 1H), 5.69 (d, 1H), 6.0 (d, 1H), 6.3 (br, 2H), 7.09 (d, 1H), 7.3-7.4(m, 7H), 7.5 (m, 3H), 7.6 (m, 1H), 7.74 (d, 2H), 8.13 (d, 2H), 8.6 (s, 1H). The mass of compound 1 was then determined by ESI-MS (positive) m/z 990.29 (M+Na)+ (Figure 16).

Synthesis of PTXL-GEM conjugate (compound 2)



[0194] Compound 1 (5 mg, 5.2 µmol) was dissolve in 0.5 mL dry DCM containing DTPS (4.6 mg, 15.6 µmol). To the solution, a solution of GEM (1.5 mg, 5.2 µmol) dissolved in 0.5 mL dry N,N-dimethylformamide (DMF) was added and solution was stirred for 15 min. After 15 min of reaction, DIPC (5 mg, 39 µmol) in 0.1 mL pyridine was added slowly to the solution and reaction was carried on at room temperature for 24 hrs. The reaction was monitored by TLC using 9.2:0.8 (v/v) CHCl3/MeOH as an eluent (product Rf = 0.22). The complete disappearance of the starting compound 1 (Rf = 0.42) occurred after 24 hrs of reaction. The reaction was then quenched by diluting the solution with dichloromethane (DCM), followed by extracting DPTS, DIPC, DMF, and pyridine with DI water. The remaining dichloromethane solution was concentrated and precipitated in hexane resulting in 6.1 mg of the compound 2 as a white powder. The production yield was about 86%. The resulting product was purified by HPLC using acetonitrile/water (50/50, v/v) as an eluent. Then 1H NMR (CDCl3, δ ppm) was carried out to characterize the produced compound 2 (Figure 10A): 0.91 (s, 1H), 1.14 (s, 3H), 1.22 (s,3H), 1.27(s,3H), 1.62 (s, 7H), 1.67 (s, 3H), 1.9-1.2 (br, 8H), 2.2-2.7 (br, 14H),2.89 (d, 2H), 3.7 (d, 2H), 3.85 (d, 2H), 3.9 (d, 1H), 4.32 (d, 1H), 4.48 (t, 1H), 5.0 (d, 1H), 5.5 (d, 1H), 5.69 (d, 1H), 6.0 (d, 1H), 6.3 (br, 3H) 7.28 (s, 3H), 7.4 (m, 5H), 7.5 (m, 3H), 7.6 (m, 1H), 7.74 (d, 2H), 8.13 (d, 2H), 8.75 (d, 1H), 9.1 (-NH2, pyrimidine ring). The mass and molecular formula of compound 2 were then determined by HR-ESI-FT-MS (orbit-trap-MS, positive) m/z 1213.4327 [M+H]+, 1235.4140 [M+Na]+. Calcd for C61H66F2N4O20 : 1213.4311. Found: 1213.4327 (Figure 10B).

Hydrolysis of PTXL-GEM conjugate (compound 2)



[0195] Hydrolysis study of PTXL-GEM conjugates was performed to confirm that the conjugates can be hydrolyzed to free PTXL and free GEM and to measure its hydrolysis kinetics at different pH values. In the study, PTXL-GEM conjugates were incubated in aqueous solutions with a pH value of 6.0 or 7.4 at 37 °C. At each predefined time interval, an aliquot of the conjugate solutions was collected and run through HPLC (mobile phase: acetonitrile/water = 50/50, v/v) to determine the amount of free PTXL, free GEM and the remaining PTXL-GEM conjugates.

Preperation of drug loaded nanoparticles



[0196] Drug loaded nanoparticles were prepared via nanoprecipitation process. In a typical experiment, 0.12 mg of lecithin (Alfa® Aesar Co.) and 0.259 mg of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-2000] (DSPE-PEG-COOH, Avinti® Polar lipids Inc.) was dissolve in 4% ethanol and homogenised to combine the components and heated at 68 °C for three minutes. To the solution 1 mg of poly(lactic-co-glycolic acid) (PLGA, Mn = 40 kDa) and calculated amount of drug dissolved in acetonitrile was added dropwise while heating and stirring. After the addition of PLGA and drug solution, the vial was vortexes for three minutes followed by the addition of 1 mL of water. The solution mixture was stirred at room temperature for 2 hrs and washed with Amicon Ultra centrifugal filter (Millipore, Billerica, MA) with a molecular weight cutoff of 10 kDa and 1 mL of drug loaded nanoparticles were collected. Bare nanoparticles were prepared similarly in the absence of drugs. The nanoparticle size and surface ξ-potential were obtained from three repeat measurements using a dynamic light scattering (Malvern Zetasizer, ZEN 3600) with backscattering angle of 173°. The morphology and particle size were further characterized using scanning electron microscopy (SEM). Samples for SEM were prepared by dropping 5 µL of nanoparticle solutions onto a polished silicon wafer. After drying the droplet at room temperature overnight, the sample was coated with chromium and then imaged by SEM. Drug loading yield was determined by using HPLC.

Cellular viability assay



[0197] Cytotoxicity of compound 2 and PTXL-GEM conjugates loaded nanoparticles was assessed against XPA3 human pancreatic carcinoma cell lines using the ATP assay. First, cells were seeded (2x104) in 96-well plates and incubated for 24 hrs. Next, the medium was replaced with 150 µL of fresh medium and incubated with different concentration of compound 2 dissolved in DMSO. The final concentration of DMSO in each well was kept constant at 2%. The plates were then incubated for 72 hrs and measured by ATP reagents following a protocol provided by the manufacturer. Fresh cell media with 2% DMSO were used as negative controls. Similar procedures were applied to compare the cytotoxicity of 100 nM of compound 2 with that of a mixture of free paclitaxel and gemcitabine at the corresponding drug concentrations at various incubation times including 24 hrs, 48 hrs, and 72 hrs. Here the use of DMSO is only for solubilizing the free drugs. For the measurement of the cytotoxicity of PTXL-GEM conjugates loaded nanoparticles, the experiments were carried out without using DMSO.

Results



[0198] Figure 9 illustrates the synthesis scheme of PTXL-GEM conjugate (compound 2). We first took advantage of the steric hindrance structural chemistry of PTXL to selectively convert its 2' hydroxyl group (2'-OH) to a carboxyl moiety (compound 1). PTXL has three hydroxyl groups, of which two are secondary and one is tertiary. It has been reported that the tertiary hydroxyl group is highly hindered and unreactive. The secondary hydroxyl group at 7 position (7-OH) is less reactive than that at 2' position. Typically, one has to protect the 2'-OH in order to make any modification to the 7-OH group. Here we used glutaric anhydride (GA) to react with PTXL in the presence of catalytic amount of N,N-dimethylaminopyridine (DMAP) for 3 hrs at room temperature

[0199] to selectively modify the 2'-OH resulting in compound 1 as characterized in Figures 14-16. We observed that the reaction had to be limited for 3 hrs with a GA:PTXL molar ratio of 3:1 for 2'-OH reaction, otherwise (longer reaction time or higher GA:PTXL ratio) 7-OH reaction occurred. Compound 1 was then reacted with GEM using 1,3-diisopropyl carbodiimide (DIPC) and 4-(N,N-dimethylamino) pyridinium-4-toluenesulfonate (DPTS) resulting in the formation of compound 2. The formation of compound 2 was first confirmed by 1H-NMR spectroscopy with all characteristic peaks and their integration values of PTXL and GEM, respectively, as indicated in Figure 10A. The 2'-OH reaction was confirmed by the integration value of 14H for the resonance peaks at δ 2.7-2.2 ppm. These peaks are corresponding to the methyl protons of acetate groups at C-4 and C-10, the methylene protons at C-14 position of the PTXL, and the methylene protons of GA linker. The resonance at δ 2.7-2.2 ppm of unmodified PTXL was integrated as 8H, which increased to 14H after the conjugation with GA because of the addition of 6H of the methylene group from GA moiety. In addition, the δ 4.4 ppm of the protons at C-7 position of PTXL remained intact during the conjugation. This further indicated the PTXL-GA reaction only occurred at the 2'-OH group as a downfield shifting of C-7 proton would have appeared if 7-OH reaction had happened. In contrast, a significant downfield shifting from δ 4.7 to δ 5.5 ppm was observed for the protons at the C-2' position. On the other hand, the use of GEM in its hydrochloride salt gives exclusive access to its hydroxyl group, which is thus prone to couple with the carboxyl group in the PTXL-GA to form an ester bond. In addition, it has been reported that DIPC and DTPS are effective esterification reagent with high reaction yield. Furthermore, the chemical shift associated with the -NH2 protons of the pyrimidine ring at 9.0 ppm were intact after the reaction. This further confirms that the PTXL-GEM conjugation occurred via ester formation. The resulting compound 2 was further examined by high resolution mass spectrometry to determine its mass and molecular formula. As shown in Figure 10B, the results were precisely consistent with the expected formula of PTXL-GEM conjugates.

[0200] As the ultimate goal of this research is to concurrently deliver dual drugs to the same cancer cells for combinatorial therapy, it is crucial to ascertain that the linker bridging the two drugs can be effectively hydrolyzed, thereby releasing individual drugs to allow them to arrest cancer cells in their independent pathways. The hydrolysis of PTXL-GEM conjugates was evaluated and confirmed by high performance liquid chromatography (HPLC) and high resolution mass spectrometry. As shown in Figure 11A, the HPLC chromatogram clearly showed that after 24 hrs of incubation in water/acetonitrile (75/25, v/v) solution at pH = 7.4, a portion of the PTXL-GEM conjugates were hydrolyzed to free PTXL and free GEM with a characteristic HPLC retention time of 6.2 min and 1.8 min, respectively, which were confirmed by measuring the mass of the compounds collected at these two retention times (see Figure 17 and 18 for the corresponding mass spectra). The formation of free PTXL and free GEM upon hydrolysis further evidenced that the PTXL-GEM conjugation occurred via the coupling of hydroxyl and carboxyl group to form an ester bond. If the reaction had occurred via amide formation between the -NH2 of the pyrimidine ring and the carboxyl group, free PTXL and free GEM would not have been released upon hydrolysis within only 24 hrs. We hypothesize that when these PTXL-GEM conjugates are delivered to target cells by a drug carrier through endocytosis, the hydrolysable PTXL-GEM conjugates can be hydrolyzed with a faster rate at the mild acidic endosomal environment (pH = ~6). To test this hypothesis, we measured the hydrolysis kinetics of the PTXL-GEM conjugates at pH = 6.0 and 7.4 respectively. As shown in Figure 11B, the hydrolysis rate was significantly faster at acidic environments (pH = 6.0) than at neutral solution (pH = 7.4). Near 80% of the drug conjugates were hydrolyzed to free PTXL and free GEM at pH = 6.0 within the first 10 hrs, while less than 25% were cleaved at pH = 7.4.

[0201] Next we examined the in vitro cellular cytotoxicity of free PTXL-GEM conjugates. As both PTXL and GEM are potent chemotherapy drugs against pancreatic cancer, we chose human pancreatic cancer cell line XPA3 for this study. Since it has been documented that the 2'-OH group is essential for high cytotoxicity of PTXL, it is natural to expect that the cytotoxicity profile of PTXL-GEM conjugates will rely on their hydrolysis process. To test this, we evaluated the cytotoxicity of the drug conjugates (100 nM concentration) at different hydrolysis duration, using a mixture of 100 nM free PTXL and 100 nM free GEM as a positive control. As shown in Figure 11C, large cytotoxicity difference was observed between the drug conjugates and the free drug mixtures after 24 hrs and 48 hrs incubation, during which the drug conjugates were partially hydrolyzed. For example, the drug conjugates killed ∼15% of XPA3 cells whereas the drug mixtures killed ∼55% of the cells after 24 hrs of incubation. However, after 72 hrs of incubation, the cytotoxicity of the PTXL-GEM conjugates was nearly at the same level as the free PTXL and free GEM mixtures; over 80% of the cells were killed for both systems. This time-dependent cytotoxicity is consistent with the temporal hydrolysis profile of the PTXL-GEM conjugates at pH = 7.4 measured by HPLC as shown in Figure 11B. It is worth noting that small molecule drugs such as PTXL, GEM and PTXL-GEM conjugate usually can diffuse across the cell membranes to the inside of the cells without going through the endocytosis mechanism. Therefore, the hydrolysis process of PTXL-GEM conjugates follows the pH = 7.4 profile when the drug conjugates are administered directly without using a drug delivery vehicle.

[0202] After having demonstrated the formation of PTXL-GEM drug conjugates, their spontaneous hydrolysis to individual drugs, and cytotoxicity against human pancreatic cancer cell line XPA3, we next loaded the PTXL-GEM conjugates into a recently developed lipid-coated polymeric nanoparticle to validate the feasibility of using this pre-conjugation approach to enable nanoparticle dual drug delivery. The PTXL-GEM conjugates were mixed with poly(lactic-co-glycolic acid) (PLGA) in an acetonitrile solution, which was subsequently added into an aqueous solution containing lipid and lipid-polyethylene glycol conjugates to prepare lipid-coated PLGA nanoparticles following a previously published protocol. L. Zhang, et al. ACS Nano 2008, 2, 1696. Figure 12A shows a schematic representation of PTXL-GEM conjugates loaded nanoparticles, which are spherical particles as imaged by scanning electron microscopy (SEM) (Figure 12B). Dynamic light scattering measurements showed that the resulting PTXL-GEM conjugates loaded nanoparticles had an unimodel size distribution with an average hydrodynamic diameter of 70 ± 1.5 nm (Figure 12C), which was consistent with the findings from the SEM image (Figure 11B). The surface zeta potential of the drug loaded nanoparticles in water was about -53 ± 2 mV (Figure 12C). We further found that the size and surface zeta potential of the PTXL-GEM conjugates loaded nanoparticles were similar to those of the corresponding empty nanoparticles, 70 ± 1 nm and -51 ± 2 mV, respectively. This suggests that the encapsulation of PTXL-GEM conjugates has negligible effect on the formation of the lipid-coated polymeric nanoparticles.

[0203] The encapsulation yield and loading yield of PTXL-GEM conjugates in the nanoparticles were quantified by HPLC after dissolving the particles in organic solvents to free all encapsulated drugs. When the initial PTXL-GEM conjugate input was 5 wt%, 10 wt%, and 15 wt% of the total nanoparticle weight, the drug encapsulation yield was 22.8 ± 2.0%, 16.2 ± 0.5%, 10.8 ± 0.7% respectively, which can be converted to the corresponding final drug loading yield of 1.1 wt%, 1.6 wt%, and 1.6 wt%, respectively (Figure 13A). Here the drug encapsulation yield is defined as the weight ratio of the encapsulated drugs to the initial drug input. The drug loading yield is defined as the weight ratio of the encapsulated drugs to the entire drug-loaded nanoparticles including both excipients and bioactive drugs. It seemed the maximum PTXL-GEM loading yield was about 1.6 wt% for the lipid-coated polymeric nanoparticles. This 1.6 wt% drug loading yield can be converted to roughly 1700 PTXL-GEM drug conjugate molecules per nanoparticle, calculating from the diameter of the nanoparticle (70 nm), PLGA density (1.2 g/mL) and the molecular weight of PTXL-GEM conjugate (1212 Da).

[0204] The cytotoxicity of PTXL-GEM conjugates loaded nanoparticles against XPA3 cell lines was then examined in comparison with free PTXL-GEM conjugates. Figure 13B summarized the results of IC50 measurements of PTXL-GEM conjugates loaded nanoparticles and free PTXL-GEM conjugates for 24 hrs incubation with the cancer cells. It was found that the IC50 value of PTXL-GEM conjugates was decreased by a factor of 200 for XPA3 cells after loading the drug conjugates into the lipid-coated polymeric nanoparticles. This enhanced cytotoxicity of PTXL-GEM conjugates upon nanoparticle encapsulation can be explained, at least partially, by the fact that nanoparticle drug delivery can suppress cancer drug resistance. Small molecule chemotherapy drugs that enter cells through either passive diffusion or membrane translocators are rapidly vacuumed out of the cells before they can take an effect by transmembrane drug efflux pumps such as P-glycoprotein (P-gp). Drug loaded nanoparticles, however, can partially bypass the efflux pumps as they are internalized through endocytosis. Once being engulfed by the plasma membrane, nanoparticles are transported by endosomal vesicles before unloading their drug payloads. Thus drug molecules are released farther away from the membrane-bound drug efflux pumps and therefore are more likely to reach and interact with their targets. The endocytic uptake mechanism is particularly favourable to the combinatorial drug delivery system present in this study. The pH drop upon the endosomal maturation into lysosomes will subject the drug conjugates to more acidic environment and more hydrolase enzymes, which will facilitate the cleavage of the hydrolysable linkers. Moreover, the degradation of PLGA polymer will also contribute to lowering the pH value surrounding the nanoparticles which can accelerate the hydrolysis process of the drug conjugates as well. The enhanced hydrolysis of the conjugate linkers may also partially answer for the near 200-fold cytotoxicity increase of PTXL-GEM conjugates after being encapsulated into the nanoparticles.

[0205] While the focus of this article is to report a novel chemical approach to loading dual chemotherapy drugs into a single nanoparticle for combinatorial drug delivery, it would be interesting to compare the cytotoxicity of PTXL-GEM conjugates loaded nanoparticles with that of a cocktail mixture of the same type of nanoparticles containing either free PTXL or free GEM. However, the vast hydrophobicity (or solubility) difference between PTXL and GEM makes it practically undoable to load them into the same type of nanoparticles, such as the lipid-coated polymeric nanoparticles used in this study. These nanoparticles can encapsulate hydrophobic drugs such as PTXL with high encapsulation and loading yields but can barely encapsulate hydrophilic drugs such as GEM. In fact, the inability of loading different drugs to the same type of nanoparticles represents a generic challenge to many pairs of drugs for combination therapy. The work presented in this paper may offer a new way to overcome this challenge.

Conclusions



[0206] In conclusion, we have demonstrated the conjugation of PTXL and GEM with a stoichiometric ratio of 1:1 via a hydrolysable ester linker and subsequently loaded the drug conjugates into lipid-coated polymeric nanoparticles. The cytotoxicity of the resulting combinatorial drug conjugates against human cancer cells was comparable to the corresponding free PTXL and GEM drug mixtures after the conjugates were hydrolyzed. The cytotoxicity of the drug conjugates was significantly improved after being encapsulated into drug delivery nanoparticles. This work provides a new method to load dual drugs to the same drug delivery vehicle in a precisely controllable manner, which holds great promise to suppress cancer drug resistance. Similar strategy may be generalized to other drug combinations. Synthesizing combinatorial drug conjugates with a broad range of stoichiometric ratios is described above.

Synthesis of Ptxl-Pt(IV) drug conjugates loaded nanoparticles



[0207] Paclitaxel and cisplatin were purchased from ChemiTek Industries Co. (SX, China) and Sigma-Aldrich Company (St. Louis, MO, USA), respectively, and used without further purification. All other materials including solvents were purchased from Sigma-Aldrich Company, USA. Single addition luminescence ATP detection assay was purchased from PerkinElmer Inc. for cytotoxicity measurement. 1H NMR spectra were recorded in CDCl3 using a Varian Mercury 500 MHz spectrometer. Electrospray ionization mass spectrometry (ESI-MS, Thermo LCQdeca mass spectrometer) and Thermo Fisher Scientific LTQ-XL Orbitrap mass spectrometer were used to determine the mass and molecular formula of the compounds. Reversed phase high performance liquid chromatography (HPLC) purification was performed on an Varian HPLC system equipped with µ-bonapack C18 column (4.6 mm×150 mm, Waters Associates, Inc.) using acetonitrile and water (50/50, v/v) as mobile phase.

Synthesis of cis,trans,cis-PtCl2(OCOCH2CH2CH2COOH)2(NH3)2 prodrug.



[0208] cis,trans,cis-PtCl2(OH)2(NH3)2 was first synthesized following a previously published protocol, (R. Kuroda, et al. X-ray and NMR studies of trans-dihydroxo-platinum(IV) antitumor complexes, J Inorg Biochem 22 (1984) 103-17; M.D. Hall, et al. The cellular distribution and oxidation state of platinum(II) and platinum(IV) antitumour complexes in cancer cells, J Biol Inorg Chem 8 (2003) 726-32) which was then used to prepare cis,trans,cis-PtCl2(OCOCH2CH2CH2 COOH)2(NH3)2. Briefly, an excess of glutaric anhydride was added to an methylene chloride (MC) solution containing 100 mg (0.3 mmol) of PtCl2(OH)2(NH3)2 under reflux condition in the presence of catalytic amount of triethylamine (TEA). After 12 h of reaction, cold water was added to hydrolyze excess glutaric anhydride. The reaction mixture was kept at 2°C for 16 hrs. The MC was then removed from the reaction mixture under reduced pressure resulting in a white residue. The residue was purified by washing with water, ethanol, and ether in that order. The final production yield was about 45%. The mass and molecular formula of cis,trans,cis-PtCl2(OCOCH2CH2CH2COOH)2(NH3)2 were then determined by HR-ESI-FT-MS (orbit-trap-MS, negative) m/z 560.97 [M-H]-, 596.83 [M+Cl]+. Calcd for C10H20Cl2N2O8Pt : 561.02. Found: 561.97 (see Figure 24).

Synthesis of Ptxl-Pt(IV) conjugate.



[0209] cis,trans,cis-PtCl2(OH)2(NH3)2 (10 µmol) and Ptxl (6 µmol) were dissolved in 200 µL dry MC. N, N-dimethylaminopyridine (DMAP, 0.57 µmol) and N, N-dicyclohexylcarbodiimide (DCC, 50 µmol) dissolved in 100 µL of dry MC were then added to this solution. The mixture solution was stirred at room temperature for 24 h. The reaction was monitored by HPLC using 50/50 (v/v) acetonitrile/water as an eluent (product retention time = 4.5 min). The complete disappearance of the starting paclitaxel (retention time = 5.5 min) occurred after 24 h of reaction. Solvent was concentrated and the byproduct dicyclohexylurea (DCU) was removed by filtration. The remaining solvent was completely removed and the residue was suspended in ethyl acetate and kept at 4 °C, during the process additional DCU precipitates out to form crystals which were further removed by filtration. The washing process was repeated three times to completely remove DCU. Finally, Ptxl-Pt(IV) conjugate was precipitated in hexane to obtain yellowish white powder. The final product was purified by HPLC with a recovery yield of 55%. 1H NMR (CDCl3, δ ppm) was carried out to characterize the produced Ptxl-Pt(IV) conjugate: 1.14 (s, 3H), 1.25 (s, 3H), 1.69 (s, 3H), 1.7-2.06 (broad, 9H), 2.16-2.27 (br, 4H), 2.3-2.7 (br, 9H), 2.9 (d, 1H), 3.2-3.6 (br,14H), 4.32 (d, 1H), 4.48 (t, 1H), 5.0 (d, 1H), 5.5 (d, 1H), 5.69 (d, 1H), 6.2-6.3 (br, 2H), 7.09 (d, 1H), 7.3-7.5(m, 10H), 8.13 (d, 2H), 8.6 (-NH), 11.0 (-COOH). The mass and molecular formula of Ptxl-Pt(IV) conjugate were determined by HR-ESI-FT-MS (orbit-trap-MS, negative) m/z 1395.32 [M-H]-, Calcd for C57H69Cl2N3O21Pt : 1396.34. Found: 1396.32.

Preparation and characterization of Ptxl-Pt(IV) drug conjugates loaded nanoparticles.



[0210] Ptxl-Pt(IV) conjugates were loaded into lipid-coated polymeric nanoparticles through a nanoprecipitation process. Typically, 0.12 mg of lecithin (Alfa® Aesar Co.) and 0.259 mg of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-2000] (DSPE-PEG-COOH, Avinti® Polar lipids Inc.) were dissolved in 4% ethanol aqueous solution and heated at 68 °C for three minutes. Then 1 mg of poly(lactic-co-glycolic acid) (PLGA, Mn=40 kDa) and calculated amount of Ptxl-Pt(IV) conjugates dissolved in acetonitrile were added drop-wise into the lipid solution under heating and stirring. After the addition of PLGA and Ptxl-Pt(IV) conjugate solution, the mixture was vortexed for 3 min followed by the addition of 1 mL of water. The resulting solution was stirred at room temperature for 2 h and washed with Amicon Ultra centrifugal filter (Millipore, Billerica, MA) with a molecular weight cutoff of 10 kDa. Finally, 1 mL of Ptxl-Pt(IV) conjugates loaded nanoparticles were collected. The nanoparticle size was obtained from three repeat measurements using a dynamic light scattering (Malvern Zetasizer, ZEN 3600) with backscattering angle of 173°. The morphology and particle size were further characterized using scanning electron microscopy (SEM). Samples for SEM were prepared by dropping 5 µL of nanoparticle solutions onto a polished silicon wafer. After drying the droplet at room temperature overnight, the sample was coated with chromium and then imaged by SEM. Drug loading yield of the nanoparticles was determined by using HPLC.

Cellular viability assay.



[0211] Cytotoxicity of free Ptxl-Pt(IV) conjugates and Ptxl-Pt(IV) conjugates loaded nanoparticles were assessed against A2780 ovarian carcinoma cell lines using the ATP assay. First, cells were seeded to 10% confluency (∼5x103/well) in 96-well plates and incubated for 24 h. Prior to the experiment, the culture medium was replaced with 150 µL fresh medium and cells were incubated with different concentration of free Ptxl-Pt(IV) conjugates and Ptxl-Pt(IV) conjugates loaded nanoparticles for 24 h, followed by washing the cells with PBS to remove excess drugs or nanoparticles. The cells were then incubated in fresh medium for 72 h and measured by ATP assay following a protocol provided by the manufacturer. Fresh culture medium was used as a negative control in this study.

Results



[0212] Figure 19 illustrates the synthesis scheme of Ptxl-Pt(IV) conjugate. We started the synthesis with the oxidation of cisplatin to form dihydroxy cisplatin, a Pt(IV) prodrug, which was later conjugated to Ptxl via a glutaric acid linker. In order to conjugate dihydroxy cisplatin with Ptxl, one can choose to first activate dihydroxy cisplatin with glutaric anhydride, followed by conjugating the resulting organo platinum complex to Ptxl. Alternatively, the conjugation can be carried out in a reverse order, where glutaric anhydride is reacted with Ptxl first and then conjugated to dihydroxy cisplatin. The difference between these two synthetic routes is that the former involves the conjugation of an organic compound with an organo platinum complex, while the latter involves a reaction between an organic compound with a dihydroxy platinum complex. Given the high flexibility to select proper reaction solvent for an organo platinum complex and Ptxl as compared to a dihydroxy platinum complex and Ptxl, we chose the first route to synthesize Ptxl-Pt(IV) hydrophobic-hydrophilic drug conjugates as shown in Figure 19.

[0213] As discussed in previous paragraph we converted Pt(IV) complex to Carboxyl functionalized organo Pt complex by reacting with GA (Supporting information Figure 24). Taking an advantage of the steric hindrance structural chemistry of Ptxl, we selectively reacted its 2' hydroxyl group (2'-OH) to a carboxyl moiety of Pt(IV) organo Pt complex. Among three -OH groups in Ptxl, it has been reported that the tertiary hydroxyl group is highly hindered and unreactive. The secondary hydroxyl group at 7 position (7-OH) is less reactive than that at 2' position. Typically, one has to protect the 2'-OH in order to make any modification to the 7-OH group.

[0214] The formation of Ptxl-Pt(IV) hydrophobic and hydrophilic conjugate was first confirmed by 1H-NMR spectroscopy with all characteristic peaks and their integration values of Ptxl and Pt(IV), respectively, as indicated in Figure 20A. The reaction at 2'-OH was confirmed due to the significant downfield shifting of the protons at the C-2' from δ 4.7 to δ 5.7 ppm. This shifting further confirms esterification between Ptxl and GA functionalized Pt(IV) thereby confirming the conjugation of Ptxl and Pt(IV) with hydrolysable linker. However, the protons at C-7 position of Ptxl remained intact at δ 4.4 ppm during the conjugation, this further indicated the Ptxl-Pt(IV) reaction only occurred at the 2'-OH group as a downfield shifting of C-7 proton would have appeared if 7-OH reaction had happened. The resulting compound 2 was further examined by high resolution mass spectroscopy to determine its mass and molecular formula. As shown in Figure 20B, the results were consistent with the expected formula of Ptxl-Pt(IV) conjugate. However, one might expect two molecules of Ptxl to attach to GA functionalized Pt(IV) due to presence of two -COOH group. Such conjugation was not observed likely because one of the GA moiety at the axial position became sterically hindered after one Ptxl was attached. The 1:1 conjugation was further confirmed by the corresponding molecular formula for Ptxl-Pt(IV) and the appearance of -COOH proton resonance at δ 11.0 ppm.

[0215] Upon completion of the conjugate synthesis and characterization, the Ptxl-Pt(IV) compound was subsequently loaded into a recently developed lipid-coated polymeric nanoparticleas demonstrated in Figure 21A to confirm whether co-encapsulation of hydrophobic and hydrophilic drugs can be accomplished using this pre-conjugation approach. Based on a previously published protocol (L. Zhang, et al. Self-assembled lipid--polymer hybrid nanoparticles: a robust drug delivery platform, ACS Nano 2 (2008) 1696-702), the Ptxl-Pt(IV) conjugates were mixed with poly(lactic-co-glycolic acid) (PLGA, Mn=40,000) in an acetonitrile solution, which was then added drop-wise in aqueous solution containing lipid and lipid-polyethylene glycol conjugates to prepare lipid-coated PLGA nanoparticles (Figure 21A). To quantify the loading yield of Ptxl-Pt(IV) conjugates, the nanoparticles were dissolved in organic solvents to free all encapsulated drugs. The solution was then analyzed by high performance liquid chromatography (HPLC). An initial Ptxl-Pt(IV) conjugate input of 10 wt% of the total polymeric nanoparticle weight yielded a final loading of 1.86% (wt/wt), or 18.6µg per 1mg of polymer (Figure 24), which is comparable with published data on nanoparticle drug loading (J.M. Chan, et al. PLGA-lecithin-PEG core-shell nanoparticles for controlled drug delivery, Biomaterials 30 (2009) 1627-34). Figure 21A shows a schematic representation of Ptxl-Pt(IV) conjugate loaded nanoparticles, which are spherical particles with unimodal size distribution with an average hydrodynamic diameter of 70 nm and a PDI of 0.21 as shown by dynamic light scattering (DLS) measurements (Figure 21B). SEM images further showed that the resulting Ptxl-Pt(IV) conjugates loaded nanoparticles had an unimodal size distribution with an average diameter of 70 nm (Figure 21C), which was consistent with the findings from DLS (Figure 21B).

[0216] After having demonstrated the loading of Ptxl-Pt(IV) conjugate, we next evaluated the in-vitro cellular cytotoxicity of Ptxl-Pt(IV) against A2780 human ovarian cancer cells as shown in Figure 22. The cells were incubated with free Pxtl-Pt(IV) conjugates and Pxtl-Pt(IV) conjugates in nanoparticles at different concentrations for 4hrs followed by PBS washing and incubation in fresh media for 72hrs before ATP cell viability assay (Figure 22A). It was observed that the Ptxl-Pt(IV) showed less toxicity as compared to that of Ptxl-Pt(IV) loaded nanoparticles. This reduced toxicity could be attributed to several factors. Firstly, the conjugation of a hydrophobic Ptxl and a hydrophilic Cisplatin gives rise to a large amphiphilic molecule that is structurally similar to phospholipids. The amphiphilic conjugate is more likely to be anchored in the lipid bilayer, resulting in less efficient drug delivery. Secondly, the cytoplasmic pH of cancer cells, which is approximately 6.8 to 7.1, cannot efficiently break the ester bond that connects the two drug molecules. In the conjugate form Ptxl and Pt(IV) cannot freely interact with their molecular targets. Therefore a slow hydrolysis rate will significantly compromise the conjugate's potency.

[0217] Cytotoxicity of the Pxtl-Pt(IV) conjugate-loaded nanoparticles provides evidence that both membrane diffusion and conjugate hydrolysis issues can be overcome by nanoparticle delivery. As shown in Figure 22A, large toxicity difference was observed between the free Ptxl-Pt(IV) and Ptxl-Pt(IV) loaded NPs system. Such difference can be easily observed from the microscopic images of the cells after the treatment with free Ptxl-Pt(IV) and Ptxl-Pt(IV) loaded NPs as shown in Figure 22B and C, respectively. The number of viable cells were significantly reduced after the treatment with Ptxl-Pt(IV) loaded NPs, Figure 2C. It has been well studied that nanoparticles below 100nm in size are taken up by cells through endocytic uptake. Upon contact with the nanoparticles the cell membranes fold inward and engulf the particles in endocytic vesicles. This process allows the drug conjugates to efficiently enter the cytoplasm without relying on passive diffusion through the lipid bilayers, which is highly unfavorable to large amphiphilic molecules. Another benefit of the endocytic uptake mechanism is that the endo-lysomal environments provides a more acidic medium which can accelerate the hydrolysis of the ester linker in the Pxtl-Cisplatin conjugate. As endosomes matures into lysosomes, their pH can drop to ∼5.5. The excess protons speed up the drug release that unblocks the functional 2'-OH of the Ptxl and relieves the Pt(IV) which reduced to Cisplatin in intracellular environment. In addition, the degradation of the PLGA polymers into lactic acid will further lower the pH value surrounding the nanoparticles, resulting in even faster drug release. The enhanced toxicity in the nanoparticle formulation of the Pxtl-Pt(IV) has significant implications as it addresses common issues in drug conjugates. Additionally, the strategy adds applicability to the fast-growing nanoparticle platforms and could potentially address the side effects associated with premature drug release in the circulation as the drug conjugates are much less potent without the vehicle.

Conclusions



[0218] In conclusion, we have demonstrated the conjugation of hydrophobic Ptxl and hydrophilic cisplatin with a hydrolysable ester linker and subsequently encapsulated the compound into a lipid-coated polymeric nanoparticle. The cytotoxicity of the resulting Ptxl-Pt(IV) conjugates against ovarian cancer cells was compared to the corresponding free Ptxl and cisplatin drug mixtures after the conjugates were hydrolyzed. The efficacy of Ptxl-Pt(IV) was significantly improved after being encapsulated into drug delivery nanoparticles. This work provides a new approach to load hydrophobic and hydrophilic drug to the same drug delivery vehicle without adding complexity to the nanoparticle structure. We demonstrate that prodrug conjugates and nanoparticulate systems can complement each other as an excellent combinatorial drug delivery platform.
TABLE 2 - Exemplary Cancers and Tumors
  adenocarcinoma, pancreatic
  adenocarcinoma, papillary, bladder
ackerman tumor adenocarcinoma, pleomorphic
adenocarcinoid, malignant, appendiceal adenocarcinoma, polymorphous low-grade
adenocarcinoma variant, gastric cancer adenocarcinoma, proximal jejunum
adenocarcinoma, alpha-fetoprotein-producing, esophageal adenocarcinoma, rete testis
adenocarcinoma, apocrine adenocarcinoma, small bowel
adenocarcinoma, appendiceal adenocarcinoma, thymus
adenocarcinoma, bartholin gland adenocarcinoma, unknown primary site
adenocarcinoma, bladder adenocarcinoma, urachal
adenocarcinoma, clear cell adenocarcinoma, urethral
adenocarcinoma, colloid adenocarcinoma, vaginal
adenocarcinoma, ductal type adenomyoepithelioma, malignant, breast
adenocarcinoma, eccrine adenosarcoma, Müllerian
adenocarcinoma, endometrioid primary, in colorectal endometriosis adrenogenital syndrome /testicular tumor
  ameloblastoma, desmoplastic
adenocarcinoma, esophagus ameloblastoma, malignant
adenocarcinoma, fallopian tube amyloid
adenocarcinoma, fetal pulmonary angioblastoma, giant cell
adenocarcinoma, gall bladder angioendothelioma, malignant, endovascular papillary
adenocarcinoma, hepatoid angioendotheliomatosis, malignant
adenocarcinoma, in situ, cervix angiomyxoma, malignant, aggressive, scrotum
adenocarcinoma, intra-extrahepatic, bile ducts angiomyxoma, malignant, aggressive, scrotum
adenocarcinoma, lacrimal gland angiosarcoma
adenocarcinoma, large bowel angiosarcoma, cardiac
adenocarcinoma, low-grade, extraosseous endolymphatic sac angiosarcoma, pulmonary artery
adenocarcinoma, mucinous angiosarcoma, Wilson-Jones
adenocarcinoma, mucinous, prostate askin tumor
adenocarcinoma, mucinous, stomach astroblastoma
adenocarcinoma, oncocytic astrocytic neoplasm
astrocytoma, anaplastic carcinoma, adenosquamous
astrocytoma, gemistocytic carcinoma, adenosquamous, liver
astrocytoma, pilocytic carcinoma, adenosquamous, pancreatic
astrocytoma, thalamic glioma carcinoma, adrenocortical
blastoma, pleuropulmonary (PPB) carcinoma, ameloblastic
blastoma, pulmonary carcinoma, anal
borderline tumor, malignant, ovary carcinoma, anaplastic
Buschke-Lowenstein tumor giant condyloma carcinoma, anaplastic, thymic
calcifying epithelial odontogenic tumor (CEOT) carcinoma, anaplastic, thyroid
carcinamitosis, peritoneal carcinoma, apocrine
carcinoid, malignant carcinoma, basal cell, perianal
carcinoid, malignant, atypical carcinoma, basal cell, vulva
carcinoid, malignant, bronchopulmonary, atypical carcinoma, basaloid squamous cell, esophageal
carcinoid, malignant, bronchopulmonary, typical carcinoma, basaloid squamous cell, NOS
carcinoid, malignant, colorectal carcinoma, basaloid, lung
carcinoid, malignant, gastric carcinoma, bile duct
carcinoid, malignant, gastrointestinal, appendix carcinoma, biliary tract
carcinoid, malignant, goblet cell carcinoma, bronchioalveolar (BAC)
carcinoid, malignant, lung carcinoma, bronchogenic small cell undifferentiated
carcinoid, malignant, pulmonary carcinoma, choroid plexus
carcinoid, malignant, rectal carcinoma, ciliated cell
carcinoid, malignant, renal carcinoma, clear cell, bladder
carcinoid, malignant, small bowel carcinoma, clear cell, eccrine
carcinoid, malignant, thymic carcinoma, clear cell, odontogenic
carcinoma, acinar cell (ACC) carcinoma, clear cell, thymic
carcinoma, acinic cell carcinoma, collecting duct (CDC)
carcinoma, adenoid basal, uterine cervix carcinoma, collecting duct, kidney
carcinoma, adenoid cystic (AdCC) carcinoma, cribriform
carcinoma, adenoid cystic, breast (ACCB) carcinoma, cribriform, breast
carcinoma, adenoid cystic, breast, metastatic (ACC-M) carcinoma, cystic
carcinoma, duodenal carcinoma, papillary, breast
carcinoma, epithelial-myoepithelial (EMC) carcinoma, parathyroid
carcinoma, gall bladder carcinoma, parietal cell
carcinoma, giant cell carcinoma, penile
carcinoma, hepatocellular carcinoma, pilomatrix
carcinoma, Hurthle cell carcinoma, pituitary
carcinoma, Hurthle cell, thyroid carcinoma, plasmacytoid urothelial, bladder
carcinoma, insular carcinoma, poorly differentiated, neuroendocrine (PDNEC)
carcinoma, insular, thyroid carcinoma, primary intraosseous
carcinoma, islet cell carcinoma, primary peritoneal, extra-ovarian (EOPPC)
carcinoma, large cell, neuroendocrine (LCNEC) carcinoma, renal cell (RCC), poorly differentiated
carcinoma, lymphoepithelioma-like, thymic carcinoma, renal cell (RCC), chromophobic (ChC)
carcinoma, male breast carcinoma, renal cell (RCC), clear cell (CCC)
carcinoma, medullary thyroid carcinoma, renal cell (RCC), collecting duct (CDC)
carcinoma, meibomian carcinoma, renal cell (RCC), papillary (PC)
carcinoma, merkel cell (MCC) carcinoma, renal cell (RCC), sarcomatoid
carcinoma, metaplastic, breast carcinoma, sarcomatoid, colon
carcinoma, microcystic adnexal carcinoma, sarcomatoid, thymic
carcinoma, mixed acinar, endocrine carcinoma, sebaceous
carcinoma, moderately differentiated, neuroendocrine carcinoma, serous ovarian, papillary(PsOC)
carcinoma, mucinous, bronchioloalveolar, lung carcinoma, signet-ring cell
carcinoma, mucinous, eccrine carcinoma, small cell
carcinoma, mucoepidermoid carcinoma, small cell undifferentiated, prostate
carcinoma, mucoepidermoid, bronchus carcinoma, small cell undifferentiated, prostrate (SCUUP)
carcinoma, nasopharyngeal/caucasians (NPC) carcinoma, small cell, anorectal neuroendocrine
carcinoma, neuroendocrine carcinoma, small cell, colorectal
carcinoma, neuroendocrine, lung carcinoma, small cell, esophageal
carcinoma, non-small cell w/neuroendocrine features, lung carcinoma, small cell, extrapulmonary
carcinoma, odontogenic carcinoma, small cell, gastrointestinal tract
carcinoma, papillary carcinoma, small cell, neuroendocrine (oat cell) (SCNC)
carcinoma, small cell, pancreatic carcinoma, tubal
carcinoma, small cell, renal carcinoma, tubular, breast
carcinoma, small cell, stomach carcinoma, undifferentiated, nasopharyngeal type (UCNT)
carcinoma, small cell, thymic carcinoma, undifferentiated, primary sinonasal nasopharyngea
carcinoma, small intestine carcinoma, undifferentiated, sinonasal (SNUC)
carcinoma, squamous cell, adnexal ductal cyst carcinoma, undifferentiated, thymic
carcinoma, squamous cell, atypical carcinoma, undifferentiated, w/lymphoid stroma
carcinoma, squamous cell, breast carcinoma, vaginal
carcinoma, squamous cell, diffuse pagetoid, esophagus carcinoma, verrucous
carcinoma, squamous cell, esophageal carcinoma, w/ spindle cell metaplasia, breast
carcinoma, squamous cell, keratinizing, thymic (KTSC) carcinoma, w/metaplasia, osteo-chondroid variant, breast
carcinoma, squamous cell, laryngeal carcinoma, w/sarcomatous metaplasia, breast
carcinoma, squamous cell, lymphoepithelioma-like carcinoma, well differentiated, neuroendocrine (WDNEC)
carcinoma, squamous cell, nasopharynx carcinoma, well differentiated, thymic (WDTC)
carcinoma, squamous cell, nonkeratinizing carcinosarcoma
carcinoma, squamous cell, oral cavity carcinosarcoma, uterine
carcinoma, squamous cell, ovarian cartilage tumor
carcinoma, squamous cell, stomach cartilaginous tumor, larynx
carcinoma, squamous cell, subungual (SCC) chemodectoma, malignant
carcinoma, squamous cell, thymic chloroma
carcinoma, squamous cell, thyroglossal duct cyst (TGDC) cholangio-carcinoma
carcinoma, squamous cell, thyroid cholangitis, primary sclerosing
carcinoma, squamous cell, urethra chondroblastoma
carcinoma, squamous cell, vagina chondroid syringoma, malignant (MCS)
carcinoma, squamous cell, vulvar chondroma, malignant, pulmonary (in Carney's triad)
carcinoma, terminal duct chondrosarcoma
carcinoma, testicular chondrosarcoma, acral synovial
carcinoma, transitional cell chondrosarcoma, classic, primary intradural
carcinoma, transitional cell, prostate chondrosarcoma, clear cell
carcinoma, trichilemmal chondrosarcoma, clear cell, larynx
chondrosarcoma, dural-based dermatofibrosarcoma protuberans, fibrosarcomatous variant
chondrosarcoma, intracranial dermatofibrosarcoma protuberans, NOS
chondrosarcoma, mesenchymal dermatofibrosarcoma protuberans, pigmented
chondrosarcoma, mysoid, extraskeletal desmoplastic, small round cell (DSRCT)
chordoma dysembryoplastic neuroepithelial tumor (DNT)
chordoma, clivus dysgerminoma
chordoma, familial dysgerminoma, ovarian
chordoma, intracranial cavity eccrine poroma, malignant
chordoma, NOS eccrine spiradenoma, malignant
chordoma, perifericum ectomesenchymoma, malignant
chordoma, sacrum emlanoma, malignant, placenta
chordoma, skull base endocrine tumor, pancreatic
chordoma, vertebrae endodermal sinus tumor
choriocarcinoma endometrioid tumor, ovary
choriocarcinoma, esophagus ependymoma
choriocarcinoma, gastric epithelial cancer, ovarian (EOC)
choriocarcinoma, ovary epithelial tumor, appendiceal
choriocarcinoma, stomach epithelial tumor, oral cavity
choriocarcinoma/male, primary, pulmonary epithelioma cuniculatum
cutaneous malignant tumor erythroleukemia
cylindroma, malignant esthesioneuroblastoma
cylindroma, malignant, apocrine fibrosarcoma
cystadenocarcinoma, acinar cell fibrous histiocytoma, malignant
cystadenocarcinoma, mucinous fibrous histiocytoma, malignant (MFH)
cystadenocarcinoma, pancreatic fibrous histiocytoma, malignant, angiomatoid
cystadenocarcinoma, serous fibrous histiocytoma, malignant, intracerebral
cystic-pseudopapillary tumor/ pancreas fibrous histiocytoma, malignant, renal
cystosarcoma phyllodes, malignant, breast fibrous tissue tumor, malignant
cystosarcoma phylloides fibrous tumor, solitary, malignant
dermatofibrosarcoma protuberans (DFSP) fibroxanthoma, atypical
follicular tumor hemangiosarcoma
ganglioneuroblastoma hepatoblastoma
gastrointestinal autonomic nerve tumor hereditary non-polyposis colorectal cancer (HNPCC)
germ cell tumor hidradenoma papilliferum, malignant
germ cell tumor, intracranial (GCTs) histiocytoma
germ cell tumor, ovarian histiocytosis, malignant
germ cell tumor, testicular (GCTS) Hodgkin's disease
germinoma (seminoma) Hodgkin's disease, bladder
germinoma, pineal Hodgkin's disease, blood
gestational trophoblastic tumor Hodgkin's disease, bone
giant cell tumor, nonendocrine Hodgkin's disease, bone marrow
glioblastoma multiforme, spinal chord Hodgkin's disease, breast
glioblastoma, giant cell Hodgkin's disease, cardiovascular system
glioma Hodgkin's disease, central nervous system
glioma, optic nerve Hodgkin's disease, connective tissue disease
glomangiosarcoma Hodgkin's disease, endocrine system
glomus tumor, malignant Hodgkin's disease, gastrointestinal tract
glucagonoma syndrome Hodgkin's disease, genitourinary
granular cell tumor, malignant Hodgkin's disease, head & neck
granular cell tumor, malignant, larynx Hodgkin's disease, kidney
granulosa cell tumor, ovary Hodgkin's disease, lung
granulosa tumor, stromal cell Hodgkin's disease, muscle
gynandroblastoma Hodgkin's disease, neurological system
hamartoma, mesenchymal, liver (MHL) Hodgkin's disease, prostate
hemangioendothelioma Hodgkin's disease, reproductive system
hemangioendothelioma, epithelioid Hodgkin's disease, respiratory system
hemangioendothelioma, spindle cell Hodgkin's disease, skin
hemangioendothelioma, thyroid Hodgkin's disease, testis
hemangioendotheliomas, epithelioid, pulmonary (PEH) Hodgkin's disease, thymus
hemangiopericytoma (HEPC) Hodgkin's disease, thyroid
hypokalemia & achlorhydria syndrome, well differentiated leukemia, acute undifferentiated (AUL)
inflammatory myofibroblastic tumor (IMT) leukemia, adult T-cell
inflammatory myofibroblastic tumor (IMT), pulmonary leukemia, basophilic
insular papillary cancer, thyroid leukemia, central nervous system
insulinoma, malignant leukemia, chronic lymphocytic (CLL)
islet cell tumor, nonfunctioning leukemia, chronic myelogenous (CML)
islet cell, pancreatic leukemia, cutis
Krukenberg leukemia, eosinophilic
Langerhans Cell Histiocytosis (LCH) leukemia, extramedullary
leiomyoblastoma leukemia, hairy cell (HCL)
leiomyomatosis, intravenous leukemia, Hodgkin's cell
leiomyosarcoma leukemia, lymphoblastic, t-cell, acute (ALL)
leiomyosarcoma, adrenal leukemia, prolymphocytic, t-cell
leiomyosarcoma, epithelioid, gastric leiomyosarcoma, gastric leukemia, promyelocytic
epithelioid Leydig cell tumor (LCT)
leiomyosarcoma, esophagus lipoastrocytoma
leiomyosarcoma, lung lipoblastoma
leiomyosarcoma, oral cavity liposarcoma
leiomyosarcoma, pancreas liposarcoma, larynx
leiomyosarcoma, primary bone (PLMSB) liposarcoma, myxoid
leiomyosarcoma, renal liposarcoma, pleomorphic
leiomyosarcoma, superficial perineal liposarcoma, primary mesenteric
leiomyosarcoma, uterine liposarcoma, renal
leiomyosarcoma, vulva liposarcoma, well-differentiated
leukemia, acute erythroblastic (FAB M6) low malignant potential tumor, ovary (LMP)
leukemia, acute lymphocytic (ALL) lymphoepithelioma, parotid gland
leukemia, acute monocytic lymphoma, adrenal
leukemia, acute myeloid (AML) lymphoma, angiocentric
leukemia, acute nonlymphocytic (ANLL) lymphoma, angiotropic large cell
leukemia, acute nonlymphoblastic lymphoma, B-cell
lymphoma, B-cell, low grade, liver lymphoma, large cell, anaplastic
lymphoma, B-cell, salivary gland lymphoma, larynx
lymphoma, bladder lymphoma, lung
lymphoma, bone lymphoma, lymphoblastic (LBL)
lymphoma, breast lymphoma, MALT
lymphoma, breast, MALT-type lymphoma, mantle cell
lymphoma, Burkitt's lymphoma, mediterranean
lymphoma, cardiovascular system lymphoma, muscle
lymphoma, central nervous system lymphoma, nasal
lymphoma, cervix lymphoma, neurological system
lymphoma, chest wall lymphoma, non-Hodgkin's (NHL)
lymphoma, colorectal mucosa associated lymphoid tumor lymphoma, non-Hodgkin's, breast
lymphoma, cutaneous B cell lymphoma, non-Hodgkin's, extranodal localization
lymphoma, cutaneous T cell (CTCL) lymphoma, non-Hodgkin's, larynx
lymphoma, diffuse large cell lymphoma, non-Hodgkin's, pulmonary
lymphoma, duodenal lymphoma, non-Hodgkin's, testis
lymphoma, endocrine lymphoma, ocular
lymphoma, esophageal lymphoma, oral
lymphoma, follicular lymphoma, orbital
lymphoma, gall bladder lymphoma, ovary
lymphoma, gastrointestinal tract lymphoma, pancreatic lymphoma, pancreas
lymphoma, genital tract lymphoma, paranasal sinus
lymphoma, head & neck lymphoma, penile
lymphoma, heart lymphoma, peripheral nervous system
lymphoma, hepatobilliary lymphoma, pharynx
lymphoma, HIV-associated lymphoma, pituitary
lymphoma, intravascular lymphoma, primary breast
lymphoma, Ki-1 positive, anaplastic, large cell lymphoma, primary central nervous system
lymphoma, kidney lymphoma, primary lung
lymphoma, large bowel lymphoma, prostate
lymphoma, pulmonary melanoma, central nervous system
lymphoma, renal melanoma, cervix
lymphoma, respiratory system melanoma, choroidal
lymphoma, scrotum melanoma, conjunctival
lymphoma, skin melanoma, desmoplastic
lymphoma, small bowel melanoma, endocrine
lymphoma, small intestine melanoma, esophageal
lymphoma, soft tissue melanoma, gall bladder
lymphoma, spermatic cord melanoma, gastrointestinal tract
lymphoma, stomach melanoma, genitourinary tract
lymphoma, t-cell (CTCL) melanoma, head & neck
lymphoma, testicular melanoma, heart
lymphoma, thyroid melanoma, intraocular
lymphoma, trachea melanoma, intraoral
lymphoma, ureter melanoma, kidney
lymphoma, urethra melanoma, larynx
lymphoma, urological system melanoma, leptomeningeal
lymphoma, uterus melanoma, lung
lymphomatosis, intravascular melanoma, nasal mucosa
MALT tumor melanoma, oral cavity
medulloblastoma melanoma, osteoid forming/ osteogenic
melanoma, adrenal melanoma, ovary
melanoma, amelanotic melanoma, pancreas
melanoma, anal melanoma, paranasal sinuses
melanoma, anorectal melanoma, parathyroid
melanoma, biliary tree melanoma, penis
melanoma, bladder melanoma, pericardium
melanoma, brain melanoma, pituitary
melanoma, breast melanoma, placenta
melanoma, cardiopulmonary system melanoma, prostate
melanoma, pulmonary mixed mesodermal tumor (MMT)
melanoma, rectum mucosa-associated lymphoid tissue (MALT)
melanoma, renal pelvis Mullerian tumor, malignant mixed, fallopian tube
melanoma, sinonasal Mullerian tumor, malignant mixed, uterine cervix
melanoma, skeletal system myeloma, IgM
melanoma, small bowel myoepithelioma
melanoma, small intestine myoepithelioma, malignant, salivary gland
melanoma, spinal cord nephroblastoma
melanoma, spleen neuroblastoma
melanoma, stomach neuroectodermal tumor, renal
melanoma, testis neuroendocrine tumor, prostate
melanoma, thyroid neurofibrosarcoma
melanoma, ureter nodular hidradenoma, malignant
melanoma, urethra oligodendroglioma
melanoma, uterus oligodendroglioma, anaplastic
melanoma, vagina oligodendroglioma, low-grade
melanoma, vulva osteosarcoma
meningioma, malignant, anaplastic Paget's disease, extramammary (EMPD)
meningioma, malignant, angioblastic Paget's disease, mammary
meningioma, malignant, atypical pancreatoblastoma
meningioma, malignant, papillary paraganglioma, malignant
mesenchymal neoplasm, stromal paraganglioma, malignant, extra-adrenal
mesenchymoma paraganglioma, malignant, gangliocytic
mesoblastic nephroma paraganglioma, malignant, laryngeal
mesothelioma, malignant peripherial nerve sheath tumor, malignant (MPNST)
mesothelioma, malignant, pleura pheochromocytoma, malignant
mesothelioma, papillary phyllodes tumor, malignant, breast
mesothelioma/tunica vaginalis, malignant (MMTV) pilomatrixoma, malignant
microadenocarcinoma, pancreatic plasmacytoma, extramedullary (EMP)
mixed cell tumor, pancreatic plasmacytoma, laryngeal
plasmacytoma, solitary sarcoma, bladder
pleomorphic adenoma, malignant sarcoma, botryoides
pleomorphic xanthoastrocytoma (PXA) sarcoma, central nervous system
plexiform fibrohistiocytic tumor sarcoma, clear cell, kidney
polyembryoma sarcoma, clear cell, soft parts
polypoid glottic tumor sarcoma, dendritic cell, follicular
primary lesions, malignant, diaphragm sarcoma, endometrial stromal (ESS)
primary malignant lesions, chest wall sarcoma, epithelioid
primary malignant lesions, pleura sarcoma, Ewing's (EWS)
primary sinonasal nasopharyngeal undifferentiated (PSNPC) sarcoma, Ewing's, extraosseus (EOE)
primitive neuroectodermal tumor (PNET) sarcoma, Ewing's, primitive neuroectodermal tumor
proliferating trichilemmal tumor, malignant sarcoma, fallopian tube
pseudomyxoma peritonei, malignant (PMP) sarcoma, fibromyxoid
raniopharyngioma sarcoma, granulocytic
reticuloendothelial tumor sarcoma, interdigitating reticulum cell
retiforme hemangioendothelioma sarcoma, intracerebral
retinoblastoma sarcoma, intracranial
retinoblastoma, trilateral sarcoma, Kaposi's
rhabdoid teratoma, atypical teratoid AT/RT sarcoma, Kaposi's, intraoral
rhabdoid tumor, malignant sarcoma, kidney
rhabdomyosarcoma (RMS) sarcoma, mediastinum
rhabdomyosarcoma, orbital sarcoma, meningeal
rhabdomyosarcoma, alveolar sarcoma, neurogenic
rhabdomyosarcoma, botryoid sarcoma, ovarian
rhabdomyosarcoma, central nervous system sarcoma, pituitary
rhabdomyosarcoma, chest wall sarcoma, pleomorphic soft tissue
rhabdomyosarcoma, paratesticular (PTR) sarcoma, primary, lung
sarcoma, adult prostate gland sarcoma, primary, pulmonar (PPS)
sarcoma, adult soft tissue sarcoma, prostate
sarcoma, alveolar soft part (ASPS) sarcoma, pulmonary arterial tree
sarcoma, renal stromal cell tumor, sex cord
sarcoma, respiratory tree stromal cell, testicular
sarcoma, soft tissue stromal luteoma
sarcoma, stromal, gastrointestinal (GIST) stromal myosis, endolymphatic (ESM)
sarcoma, stromal, ovarian stromal tumor, colorectal
sarcoma, synovial stromal tumor, gastrointestinal (GIST)
sarcoma, synovial, intraarticular stromal tumor, gonadal (sex cord) (GSTS)
sarcoma, synovial, lung stromal tumor, ovary
sarcoma, true stromal tumor, small bowel
sarcoma, uterine struma ovarii
sarcoma, vaginal teratocarcinosarcoma, sinonasal (SNTCS)
sarcoma, vulvar teratoma, immature
sarcomatosis, meningeal teratoma, intramedullary spine
sarcomatous metaplasia teratoma, mature
schwannoma, malignant teratoma, pericardium
schwannoma, malignant, cellular, skin teratoma, thyroid gland
schwannoma, malignant, epithelioid thecoma stromal luteoma
schwannoma, malignant, esophagus thymoma, malignant
schwannoma, malignant, nos thymoma, malignant, medullary
Sertoli cell tumor, large cell, calcifying thyroid/brain, anaplastic
sertoli-Leydig cell tumor (SLCT) trichoblastoma, skin
small cell cancer, lungsmall cell lung cancer (SCLC) triton tumor, malignant, nasal cavity
solid-pseudopapillary tumor, pancreas trophoblastic tumor, fallopian tube
somatostinoma trophoblastic tumor, placental site
spindle cell tumor urethral cancer
spindle epithelial tumour w/thymus-like element vipoma (islet cell)
spiradenocylindroma, kidney vulvar cancer
squamous neoplasm, papillary Waldenstrom's macroglobullinemia
steroid cell tumor Wilms' tumor Nephroblastoma
Stewart-Treves syndrome Wilms' tumor, lung
TABLE 3 - Exemplary Cancer Medications
  Cabazitaxel
  Campath (Alemtuzumab)
Abiraterone Acetate Camptosar (Irinotecan Hydrochloride)
Abitrexate (Methotrexate) Capecitabine
Adriamycin (Doxorubicin Hydrochloride) Carboplatin
Adrucil (Fluorouracil) Cerubidine (Daunorubicin Hydrochloride)
Afinitor (Everolimus) Cervarix (Recombinant HPV Bivalent Vaccine)
Aldara (Imiquimod) Cetuximab
Aldesleukin Chlorambucil
Alemtuzumab Cisplatin
Alimta (Pemetrexed Disodium) Clafen (Cyclophosphamide)
Aloxi (Palonosetron Hydrochloride) Clofarabine
Ambochlorin (Chlorambucil) Clofarex (Clofarabine)
Amboclorin (Chlorambucil) Clolar (Clofarabine)
Aminolevulinic Acid Cyclophosphamide
Anastrozole Cyfos (Ifosfamide)
Aprepitant Cytarabine
Arimidex (Anastrozole) Cytarabine, Liposomal
Aromasin (Exemestane) Cytosar-U (Cytarabine)
Arranon (Nelarabine) Cytoxan (Cyclophosphamide)
Arsenic Trioxide Dacarbazine
Arzerra (Ofatumumab) Dacogen (Decitabine)
Avastin (Bevacizumab) Dasatinib
Azacitidine Daunorubicin Hydrochloride
Bendamustine Hydrochloride Decitabine
Bevacizumab Degarelix
Bexarotene Denileukin Diftitox
Bexxar (Tositumomab and I 131 Iodine Tositumomab) Denosumab
Bleomycin DepoCyt (Liposomal Cytarabine)
Bortezomib DepoFoam (Liposomal Cytarabine)
Dexrazoxane Hydrochloride Fulvestrant
Docetaxel Gardasil (Recombinant HPV Quadrivalent Vaccine)
Doxorubicin Hydrochloride Gefitinib
Efudex (Fluorouracil) Gemcitabine Hydrochloride
Elitek (Rasburicase) Gemtuzumab Ozogamicin
Ellence (Epirubicin Hydrochloride) Gemzar (Gemcitabine Hydrochloride)
Eloxatin (Oxaliplatin) Gleevec (Imatinib Mesylate)
Eltrombopag Olamine Halaven (Eribulin Mesylate)
Emend (Aprepitant) Herceptin (Trastuzumab)
Epirubicin Hydrochloride HPV Bivalent Vaccine, Recombinant
Erbitux (Cetuximab) HPV Quadrivalent Vaccine, Recombinant
Eribulin Mesylate Hycamtin (Topotecan Hydrochloride)
Erlotinib Hydrochloride Ibritumomab Tiuxetan
Etopophos (Etoposide Phosphate) Ifex (Ifosfamide)
Etoposide Ifosfamide
Etoposide Phosphate Ifosfamidum (Ifosfamide)
Everolimus Imatinib Mesylate
Evista (Raloxifene Hydrochloride) Imiquimod
Exemestane Ipilimumab
Fareston (Toremifene) Iressa (Gefitinib)
Faslodex (Fulvestrant) Irinotecan Hydrochloride
Femara (Letrozole) Istodax (Romidepsin)
Filgrastim Ixabepilone
Fludara (Fludarabine Phosphate) Ixempra (Ixabepilone)
Fludarabine Phosphate Jevtana (Cabazitaxel)
Fluoroplex (Fluorouracil) Keoxifene (Raloxifene Hydrochloride)
Fluorouracil Kepivance (Palifermin)
Folex (Methotrexate) Lapatinib Ditosylate
Folex PFS (Methotrexate) Lenalidomide
Folotyn (Pralatrexate) Letrozole
Leucovorin Calcium Ofatumumab
Leukeran (Chlorambucil) Oncaspar (Pegaspargase)
Leuprolide Acetate Ontak (Denileukin Diftitox)
Levulan (Aminolevulinic Acid) Oxaliplatin
Linfolizin (Chlorambucil) Paclitaxel
LipoDox (Doxorubicin Hydrochloride Liposome) Palifermin
Liposomal Cytarabine Palonosetron Hydrochloride
Lupron (Leuprolide Acetate) Panitumumab
Lupron Depot (Leuprolide Acetate) Paraplat (Carboplatin)
Lupron Depot-Ped (Leuprolide Acetate) Paraplatin (Carboplatin)
Lupron Depot-3 Month (Leuprolide Acetate) Pazopanib Hydrochloride
Lupron Depot-4 Month (Leuprolide Acetate) Pegaspargase
Matulane (Procarbazine Hydrochloride) Pemetrexed Disodium
Methazolastone (Temozolomide) Platinol (Cisplatin)
Methotrexate Platinol-AQ (Cisplatin)
Methotrexate LPF (Methotrexate) Plerixafor
Mexate (Methotrexate) Pralatrexate
Mexate-AQ (Methotrexate) Prednisone
Mozobil (Plerixafor) Procarbazine Hydrochloride
Mylosar (Azacitidine) Proleukin (Aldesleukin)
Mylotarg (Gemtuzumab Ozogamicin) Prolia (Denosumab)
Nanoparticle Paclitaxel (Paclitaxel Albumin-stabilized Nanoparticle Promacta (Eltrombopag Olamine)
Formulation) Provenge (Sipuleucel-T)
Nelarabine Raloxifene Hydrochloride
Neosar (Cyclophosphamide) Rasburicase
Neupogen (Filgrastim) Recombinant HPV Bivalent Vaccine
Nexavar (Sorafenib Tosylate) Recombinant HPV Quadrivalent Vaccine
Nilotinib Revlimid (Lenalidomide)
Nolvadex (Tamoxifen Citrate) Rheumatrex (Methotrexate)
Nplate (Romiplostim) Rituxan (Rituximab)
Rituximab Tositumomab and I 131 Iodine Tositumomab
Romidepsin Totect (Dexrazoxane Hydrochloride)
Romiplostim Trastuzumab
Rubidomycin (Daunorubicin Hydrochloride) Treanda (Bendamustine Hydrochloride)
Sclerosol Intrapleural Aerosol (Talc) Trisenox (Arsenic Trioxide)
Sipuleucel-T Tykerb (Lapatinib Ditosylate)
Sorafenib Tosylate Vandetanib
Sprycel (Dasatinib) Vectibix (Panitumumab)
Sterile Talc Powder (Talc) Velban (Vinblastine Sulfate)
Steritalc (Talc) Velcade (Bortezomib)
Sunitinib Malate Velsar (Vinblastine Sulfate)
Sutent (Sunitinib Malate) VePesid (Etoposide)
Synovir (Thalidomide) Viadur (Leuprolide Acetate)
Talc Vidaza (Azacitidine)
Tamoxifen Citrate Vinblastine Sulfate
Tarabine PFS (Cytarabine) Vincasar PFS (Vincristine Sulfate)
Tarceva (Erlotinib Hydrochloride) Vincristine Sulfate
Targretin (Bexarotene) Vorinostat
Tasigna (Nilotinib) Votrient (Pazopanib Hydrochloride)
Taxol (Paclitaxel) Wellcovorin (Leucovorin Calcium)
Taxotere (Docetaxel) Xeloda (Capecitabine)
Temodar (Temozolomide) Xgeva (Denosumab)
Temozolomide Yervoy (Ipilimumab)
Temsirolimus Zevalin (Ibritumomab Tiuxetan)
Thalidomide Zinecard (Dexrazoxane Hydrochloride)
Thalomid (Thalidomide) Zoledronic Acid
Toposar (Etoposide) Zolinza (Vorinostat)
Topotecan Hydrochloride Zometa (Zoledronic Acid)
Toremifene Zytiga (Abiraterone Acetate)
Torisel (Temsirolimus)  
TABLE 4 - Exemplary Ocular Diseases and Conditions
    keratoconjunctivitis sicca (dry eye syndrome)
Examples of "back of the eye" diseases include iridocyclitis
  • macular edema such as angiographic cystoid macular iritis
  edema scleritis
  • retinal ischemia and choroidal neovascularization episcleritis
  • macular degeneration corneal edema
  • retinal diseases (e.g., diabetic retinopathy, diabetic scleral disease
  retinal edema, retinal detachment); inflammatory ocular cicatrcial pemphigoid
  diseases such as uveitis (including panuveitis) or pars planitis
  choroiditis (including multifocal choroiditis) of Posner Schlossman syndrome
  unknown cause (idiopathic) or associated with a Behcet's disease
  systemic (e.g., autoimmune) disease; episcleritis or Vogt-Koyanagi-Harada syndrome
  scleritis hypersensitivity reactions
  • Birdshot retinochoroidopathy conjunctival edema
  • vascular diseases (retinal ischemia, retinal vasculitis,
  choroidal vascular insufficiency, choroidal conjunctival venous congestion
  thrombosis) periorbital cellulitis; acute dacryocystitis
  • neovascularization of the optic nerve non-specific vasculitis
  • optic neuritis sarcoidosis
Examples of "front-of-eye" diseases include:    
  • blepharitis    
  • keratitis    
  • rubeosis iritis    
  • Fuchs' heterochromic iridocyclitis    
  • chronic uveitis or anterior uveitis    
  • conjunctivitis    
  • allergic conjunctivitis (including seasonal or perennial, vernal, atopic, and giant papillary)    
TABLE 5 - Exemplary Ocular Medications
Atropine
Brimondine (Alphagan)
Ciloxan
Erythromycin
Gentamicin
Levobunolol (Betagan)
Metipranolol (Optipranolol)
Optivar
Patanol
PredForte
Proparacaine
Timoptic
Trusopt
Visudyne (Verteporfin)
Voltaren
Xalatan
TABLE 6 - Exemplary Diseases and Conditions affecting the Lungs
Acute Bronchitis Pulmonary Arterial Hypertension
Acute Respiratory Distress Syndrome (ARDS) Pulmonary Fibrosis
Asbestosis Pulmonary Vascular Disease
Asthma Respiratory Syncytial Virus
Bronchiectasis Sarcoidosis
Bronchiolitis Severe Acute Respiratory Syndrome
Bronchopulmonary Dysplasia Silicosis
Byssinosis Sleep Apnea
Chronic Bronchitis Sudden Infant Death Syndrome
Coccidioidomycosis (Cocci) Tuberculosis
COPD  
Cystic Fibrosis  
Emphysema  
Hantavirus Pulmonary Syndrome  
Histoplasmosis  
Human Metapneumovirus  
Hypersensitivity Pneumonitis  
Influenza  
Lung Cancer  
Lymphangiomatosis  
Mesothelioma  
Nontuberculosis Mycobacterium  
Pertussis  
Pneumoconiosis  
Pneumonia  
Primary Ciliary Dyskinesia  
Primary Pulmonary Hypertension  
TABLE 7 - Exemplary Lung/Respiratory disease medications:
  Curosurf
  Daliresp (roflumilast)
Accolate Dulera (mometasone furoate + formoterol fumarate dihydrate)
Accolate DuoNeb (albuterol sulfate and ipratropium bromide)
Adcirca (tadalafil) Dynabac
Aldurazyme (laronidase) Flonase Nasal Spray
Allegra (fexofenadine hydrochloride) Flovent Rotadisk
Allegra-D Foradil Aerolizer (formoterol fumarate) inhalation powder)
Alvesco (ciclesonide) Infasurf
Astelin nasal spray Invanz
Atrovent (ipratropium bromide) Iressa (gefitinib)
Augmentin (amoxicillin/clavulanate) Ketek (telithromycin)
Avelox I.V. (moxifloxacin hydrochloride) Letairis (ambrisentan)
Azmacort (triamcinolone acetonide) Inhalation Aerosol Metaprotereol Sulfate Inhalation Solution, 5%
Biaxin XL (clarithromycin extended-release tablets) Nasacort AQ (triamcinolone acetonide) Nasal Spray
Breathe Right Nasacort AQ (triamcinolone acetonide) Nasal Spray
Brovana (arformoterol tartrate) NasalCrom Nasal Spray
Cafcit Injection OcuHist
Cayston (aztreonam for inhalation solution) Omnicef
Cedax (ceftibuten) Patanase (olopatadine hydrochloride)
Cefazolin and Dextrose USP Priftin
Ceftin (cefuroxime axetil) Proventil HFA Inhalation Aerosol
Cipro (ciprofloxacin HCl) Pulmozyme (dornase alfa)
Clarinex Pulmozyme (dornase alfa)
Claritin RediTabs (10 mg loratadine rapidly-disintegrating tablet) Qvar (beclomethasone dipropionate)
Claritin Syrup (loratadine) Raxar (grepafloxacin)
Claritin-D 24 Hour Extended Release Tablets (10 mg loratadine, Remodulin (treprostinil)
240 mg pseudoephedrine sulfate) RespiGam (Respiratory Syncitial Virus Immune Globulin
Clemastine fumarate syrup Intravenous)
Covera-HS (verapamil) Rhinocort Aqua Nasal Spray
Sclerosol Intrapleural Aerosol Zosyn (sterile piperacillin sodium/tazobactam sodium)
Serevent Zyflo (Zileuton)
Singulair Zyrtec (cetirizine HCl)
Spiriva HandiHaler (tiotropium bromide)  
Synagis  
Tavist (clemastine fumarate)  
Tavist (clemastine fumarate)  
Teflaro (ceftaroline fosamil)  
Tequin  
Tikosyn Capsules  
Tilade (nedocromil sodium)  
Tilade (nedocromil sodium)  
Tilade (nedocromil sodium)  
Tobi  
Tracleer (bosentan)  
Tri-Nasal Spray (triamcinolone acetonide spray)  
Tripedia (Diptheria and Tetanus Toxoids and Acellular Pertussis  
Vaccine Absorbed)  
Tygacil (tigecycline)  
Tyvaso (treprostinil)  
Vancenase AQ 84 mcg Double Strength  
Vanceril 84 mcg Double Strength (beclomethasone dipropionate, 84  
mcg) Inhalation Aerosol  
Ventolin HFA (albuterol sulfate inhalation aerosol)  
Visipaque (iodixanol)  
Xolair (omalizumab)  
Xopenex  
Xyzal (levocetirizine dihydrochloride)  
Zagam (sparfloxacin) tablets  
Zemaira (alphal-proteinase inhibitor)  
TABLE 8 - Exemplary Diseases and Conditions affecting the Heart:
Heart attack
Atherosclerosis
High blood pressure
Ischemic heart disease
Heart rhythm disorders
Tachycardia
Heart murmurs
Rheumatic heart disease
Pulmonary heart disease
Hypertensive heart disease
Valvular heart disease
Infective endocarditis
Congenital heart diseases
Coronary heart disease
Atrial myxoma
HOCM
Long QT syndrome
Wolff Parkinson White syndrome
Supraventricular tachycardia
Atrial flutter
Constrictive pericarditis
Atrial myxoma
Long QT syndrome
Wolff Parkinson White syndrome
Supraventricular tachycardia
Atrial flutter
TABLE 9 - Exemplary Heart Medications
  nadolol, Corgard
  niacin and lovastatin, Advicor
ACE Inhibitors niacin, Niacor, Niaspan, Slo-Niacin
acetylsalicylic acid, Aspirin, Ecotrin nitroglycerin, Nitro-Bid, Nitro-Dur, Nitrostat, Transderm-
alteplase, Activase, TPA Nitro, Minitran, Deponit, Nitrol
anistreplase-injection, Eminase oxprenolol-oral
Aspirin and Antiplatelet Medications pravastatin, Pravachol
atenolol, Tenormin pravastatin/buffered aspirin-oral, Pravigard PAC
atorvastatin, Lipitor propranolol, Inderal, Inderal LA
benazepril, Lotensin quinapril hcl/hydrochlorothiazide-oral, Accuretic
Beta Blockers quinapril, Accupril
Bile Acid Sequestrants ramipril, Altace
Calcium Channel Blockers reteplase-injection, Retavase
captopril and hydrochlorothiazide, Capozide simvastatin, Zocor
captopril, Capoten Statins
clopidogrel bisulfate, Plavix streptokinase-injection, Kabikinase, Streptase
colesevelam, Welchol torsemide-oral, Demadex
dipyridamole-oral, Persantine trandolapril, Mavik
enalapril and hydrochlorothiazide, Vaseretic  
enalapril, Vasotec  
ezetimibe and simvastatin, Vytorin  
Fibrates  
fluvastatin, Lescol  
fosinopril sodium, Monopril  
lisinopril and hydrochlorothiazide, Zestoretic, Prinzide  
lisinopril, Zestril, Prinivil  
lovastatin, Mevacor, Altocor  
magnesium sulfate-injection  
metoprolol, Lopressor, Toprol XL  
moexipril-oral, Univasc  
TABLE 10 - Exemplary Bacterial, Viral, Fungal and Parasitic Conditions
Bacterial Infections caused by:
  • Borrelia species
  • Streptococcus pneumoniae
  • Staphylococcus aureus
  • Mycobacterium tuberculosis
  • Mycobacterium leprae
  • Neisseria gonorrheae
  • Chlamydia trachomatis
  • Pseudomonas aeruginosa
 
Viral Infections caused by:
  • Herpes simplex
  • Herpes zoster
  • cytomegalovirus
 
Fungal Infections caused by:
  • Aspergillus fumigatus
  • Candida albicans
  • Histoplasmosis capsulatum
  • Cryptococcus species
  • Pneumocystis carinii
 
Parasitic Infections caused by:
  • Toxoplasmosis gondii
  • Trypanosome cruzi
  • Leishmania species
  • Acanthamoeba species
  • Giardia lamblia
  • Septata species
  • Dirofilaria immitis



Claims

1. A method for nanoencapsulation of a plurality of drugs comprising:

separately linking each of the plurality of drugs with a corresponding polymer backbone with nearly 100% loading efficiency by forming the corresponding polymer backbone by ring opening polymerization beginning with the corresponding drug, wherein each of the corresponding polymer backbones has the same or similar physicochemical properties and has approximately the same chain length;

mixing the plurality of linked drugs and polymers at selectively predetermined ratios at selectively and precisely controlled drug ratios; and

synthesizing the mixed plurality of linked drugs and polymers into a nanoparticle.


 
2. The method of claim 1, wherein the plurality of drugs are independently selected from the group consisting of an antibiotic, antimicrobial, growth factor, chemotherapeutic agent, and combinations thereof.
 
3. The method of claims 1 to 2, wherein the plurality of drugs comprise an anticancer chemotherapeutic agent.
 
4. The method of claim 1 to 2, wherein the plurality of drugs are independently selected from the group consisting of doxorubicin, camptothecin, gemcitabine, carboplatin, oxaliplatin, epirubicin, idarubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloromethotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, cytosine arabinoside, podophyllotoxin, etoposide, etoposide phosphate, melphalan, vinblastine, vincristine, leurosidine, vindesine, estramustine, cisplatin, cyclophosphamide, paclitaxel, leurositte, 4-desacetylvinblastine, epothilone B, docetaxel, maytansinol, epothilone A, combretastatin, pharmaceutically active analogs thereof, and pharmaceutically acceptable salts thereof.
 
5. The method of claim 1, wherein the polymer backbone is a stimuli-sensitive linker.
 
6. The method of claim 5, wherein the linker is a pH-sensitive linker.
 
7. The method of claim 5, wherein the stimuli-sensitive linker is selected from the group consisting of C1-C10 straight chain alkyl, C1-C10 straight chain O-alkyl, C1-C10 straight chain substituted alkyl, C1-C10 straight chain substituted O-alkyl, C4-C13 branched chain alkyl, C4-C13 branched chain O-alkyl, C2-C12 straight chain alkenyl, C2-C12 straight chain O-alkenyl, C3-C12 straight chain substituted alkenyl, C3-C12 straight chain substituted O-alkenyl, polyethylene glycol, polylactic acid, polyglycolic acid, poly(lactide-co-glycolide), polycarprolactone, polycyanoacrylate, ketone, aryl, aralkyl, heterocyclic, and combinations thereof.
 
8. The method of claims 1 to 7, wherein said nanoparticle is a lipid-polymer hybrid nanoparticle with a polymeric core consisting of the plurality of linked drugs and polymers.
 
9. The method of claim 8, wherein said nanoparticle is a lipid-coated polymeric nanoparticle.
 
10. The method of claims 1 to 9, wherein the nanoparticle is about 1 nm to about 10 µm in diameter.
 
11. The method of claim 10, wherein the nanoparticle is about 30 nm to about 300 nm in diameter.
 


Ansprüche

1. Verfahren zur Nanoeinkapselung mehrerer Arzneimittel, umfassend:

separates Verbinden jedes der mehreren Arzneimittel mit einem entsprechenden Polymergerüst mit nahezu 100 % Beladungseffizienz durch Bilden des entsprechenden Polymergerüsts durch Ringöffnungspolymerisation, die mit dem entsprechenden Arzneimittel beginnt, worin jedes der entsprechenden Polymergerüste die gleichen oder ähnliche physikochemischen Eigenschaften aufweist und ungefähr die gleiche Kettenlänge aufweist;

Mischen der mehreren verbundenen Arzneimittel und Polymere in ausgewählten vorherbestimmten Verhältnissen unter ausgewählten und genau kontrollierten Arzneimittelverhältnissen; und

Synthetisieren der gemischten mehreren verbundenen Arzneimittel und Polymere in ein Nanoteilchen.


 
2. Verfahren nach Anspruch 1, worin die mehreren Arzneimittel unabhängig ausgewählt sind aus der Gruppe, bestehend aus einem Antibiotikum, antimikrobiellen Mittel, einem Wachstumsfaktor, einem chemotherapeutischen Mittel und Kombinationen davon.
 
3. Verfahren nach Anspruch 1 oder 2, worin die mehreren Arzneimittel ein Antkrebschemotherapiemittel umfassen.
 
4. Verfahren nach Anspruch 1 bis 2, worin die mehreren Arzneimittel unabhängig ausgewählt sind aus der Gruppe, bestehend aus Doxorubicin, Camptothecin, Gemcitabin, Carboplatin, Oxaliplatin, Epirubicin, Idarubicin, Carminomycin, Daunorubicin, Aminopterin, Methotrexat, Methopterin, Dichlormethotrexat, Mitomycin C, Porfiromycin, 5-Fluoruracil, 6-Mercaptopurin, Cytosinarbinosid, Podophyllotoxin, Etoposid, Etoposidphosphat, Melphalan, Vinblastin, Vincristin, Leurosidin, Vindesin, Estramustin, Cisplatin, Cyclophospamid, Paclitaxel, Leurositte, 4-Desacetylvinblastin, Epothilon B, Docetaxel, Maytansinol, Epothilon A, Combretastatin, pharmazeutisch aktiven Analogen davon und pharmazeutisch verträgliche Salze davon.
 
5. Verfahren nach Anspruch 1, worin das Polymergerüst ein Stimulisensitiver Linker ist.
 
6. Verfahren nach Anspruch 5, worin der Linker ein pH-sensitiver Linker ist.
 
7. Verfahren nach Anspruch 5, worin der Stimuli-sensitive Linker ausgewählt ist aus der Gruppe, bestehend aus geradkettigem C1-C10-Alkyl, geradkettigem C1-C10-O-Alkyl, geradkettigem substituiertem C1-C10-Alkyl, geradkettigem substituiertem C1-C10-O-Alkyl, verzweigtkettigem C4-C13-Alkyl, verzweigtkettigem C4-C13-O-Alkyl, geradkettigem C2-C12-Alkenyl, geradkettigem C2-C12-O-Alkenyl, geradkettigem substituiertem C3-C12-Alkenyl, geradkettigem substituiertem C3-C12-O-Alkenyl, Polyethylenglykol, Polymilchsäure, Polyglykolsäure, Poly(lactid-co-glycolid), Polycaprolacton, Polycyanacrylat, Keton, Aryl, Aralkyl, heterozyklisch und Kombinationen davon.
 
8. Verfahren nach den Ansprüchen 1 bis 7, worin das Nanoteilchen ein Lipid-Polymer-Hybrid-Nanoteilchen mit einem polymeren Kern, bestehend aus den mehreren verbundenen Arzneimitteln und Polymeren, ist.
 
9. Verfahren nach Anspruch 8, worin das Nanoteilchen ein Lipdbeschichtetes polymeres Nanoteilchen ist.
 
10. Verfahren nach einem der Ansprüche 1 bis 9, worin das Nanoteilchen einen Durchmesser von etwa 1 nm bis etwa 10 µm aufweist.
 
11. Verfahren nach einem der Ansprüche 1 bis 9, worin das Nanoteilchen einen Durchmesser von etwa 30 nm bis etwa 300 nm aufweist.
 


Revendications

1. Procédé pour la nanoencapsulation d'une pluralité de médicaments comprenant:

la liaison séparément de chacun de la pluralité de médicaments avec un squelette polymère correspondant avec sensiblement 100 % d'efficacité de chargement par formation du squelette polymère correspondant par polymérisation par ouverture de cycle commençant avec le médicament correspondant, où chacun des squelettes polymères correspondants a des propriétés physicochimiques identiques ou similaires et a approximativement la même longueur de chaîne;

le mélange de la pluralité de médicaments et de polymères liés à des rapports sélectivement prédéterminés à des rapports de médicaments sélectivement et précisément contrôlés; et

la synthèse de la pluralité mélangée de médicaments et de polymères liés en une nanoparticule.


 
2. Procédé selon la revendication 1 où la pluralité de médicaments est choisie indépendamment dans le groupe consistant en un antibiotique, un antimicrobien, un facteur de croissance, un agent chimiothérapeutique et leurs combinaisons.
 
3. Procédé selon les revendications 1 à 2 où la pluralité de médicaments comprend un agent chimiothérapeutique anticancéreux.
 
4. Procédé selon les revendications 1 à 2 où la pluralité de médicaments est choisie indépendamment dans le groupe consistant en la doxorubicine, la camptothécine, la gemcitabine, le carboplatine, l'oxaliplatine, l'épirubicine, l'idarubicine, la carminomycine, la daunorubicine, l'aminoptérine, le méthotrexate, la méthoptérine, le dichlorométhotrexate, la mitomycine C, la porfiromycine, le 5-fluorouracile, la 6-mercaptopurine, l'arabinoside de cytosine, la podophyllotoxine, l'étoposide, l'étoposide phosphate, le melphalan, la vinblastine, la vincristine, la leurosidine, la vindésine, l'estramustine, le cisplatine, le cyclophosphamide, le paclitaxel, la leurositte, la 4-désacétylvinblastine, l'épothilone B, le docétaxel, le maytansinol, l'épothilone A, la combrétastatine, leurs analogues pharmaceutiquement actifs et leurs sels pharmaeeutiquement acceptables.
 
5. Procédé selon la revendication 1 où le squelette polymère est un lieur sensible aux stimuli.
 
6. Procédé selon la revendication 5 où le lieur est un lieur sensible au pH.
 
7. Procédé selon la revendication 5 où le lieur sensible aux stimuli est choisi dans le groupe consistant en C1-C10 alkyle linéaire, C1-C10 O-alkyle linéaire, C1-C10 alkyle linéaire substitué, C1-C10 O-alkyle linéaire substitué, C4-C13 alkyle ramifié, C4-C13 O-alkyle ramifié, C2-C12 alcényle linéaire, C2-C12 O-alcényle linéaire, C3-C12 alcényle linéaire substitué, C3-C12 O-alcényle linéaire substitué, le polyéthylèneglycol, le poly(acide lactique), le poly(acide glycolique), un copolymère lactide-glycolide, la polycaprolactone, un polycyanoacrylate, une cétone, un aryle, aralkyle, hétérocyclique, et leurs combinaisons.
 
8. Procédé selon les revendications 1 à 7 où ladite nanoparticule est une nanoparticule hybride lipide-polymère avec un noyau polymérique consistant en la pluralité de médicaments et de polymères liés.
 
9. Procédé selon la revendication 8 où ladite nanoparticule est une nanoparticule polymérique à enrobage lipidique.
 
10. Procédé selon les revendications 1 à 9 où la nanoparticule a un diamètre d'environ 1 nm à environ 10 µm.
 
11. Procédé selon la revendication 10 où la nanoparticule a un diamètre d'environ 30 nm à environ 300 nm.
 




Drawing



















































































Cited references

REFERENCES CITED IN THE DESCRIPTION



This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.

Patent documents cited in the description




Non-patent literature cited in the description